WO2011106607A2 - Subunit vaccines for herpes viruses and methods of use - Google Patents

Subunit vaccines for herpes viruses and methods of use Download PDF

Info

Publication number
WO2011106607A2
WO2011106607A2 PCT/US2011/026188 US2011026188W WO2011106607A2 WO 2011106607 A2 WO2011106607 A2 WO 2011106607A2 US 2011026188 W US2011026188 W US 2011026188W WO 2011106607 A2 WO2011106607 A2 WO 2011106607A2
Authority
WO
WIPO (PCT)
Prior art keywords
glycoprotein
fragment
composition
nucleic acid
hsv
Prior art date
Application number
PCT/US2011/026188
Other languages
French (fr)
Other versions
WO2011106607A3 (en
Inventor
Jeffery Fairman
Gary H. Cohen
Roselyn J. Eisenberg
Original Assignee
Juvaris Biotherapeutics, Inc.
The Trustees Of The University Of Pennsylvania
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Juvaris Biotherapeutics, Inc., The Trustees Of The University Of Pennsylvania filed Critical Juvaris Biotherapeutics, Inc.
Priority to US13/578,976 priority Critical patent/US9089537B2/en
Publication of WO2011106607A2 publication Critical patent/WO2011106607A2/en
Publication of WO2011106607A3 publication Critical patent/WO2011106607A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5252Virus inactivated (killed)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16622New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16634Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Abstract

The present disclosure generally relates to vaccine compositions for Herpes Simplex Viruses (HSV) types 1 and/or 2. The vaccines comprise isolated antigens or glycoprotein subunits of the viruses, optionally with an adjuvant, such as a cationic liposome DNA complex (CLDC). Also the present disclosure contains methods of vaccinating a subject utilizing these compositions.

Description

SUBUNIT VACCINES FOR HERPES VIRUSES AND METHODS OF USE
RELATED APPLICATIONS
[0001] This application claims benefit of priority from U.S. Provisional Application
Serial No. 61/308,856, filed February 26, 2010, currently pending, the entire content of which is incorporated by reference as if fully set forth.
BACKGROUND OF THE INVENTION FIELD OF THE INVENTION
[0002] The invention relates generally to herpes simplex virus (HSV) vaccines, and more specifically to vaccine compositions of single and multi-antigen HSV subunit proteins, with or without an adjuvant, such as a cationic liposome delivery vehicle, and the uses of these compositions to immunize against herpes simplex virus types.
BACKGROUND INFORMATION
[0003] Herpes simplex viruses (HSVs) cause human diseases, including, for example, cold sores, eye and genital infections, neonatal infections and encephalitis. There are two serotypes of the virus, the oral form, termed HSV-1 and the genital form, termed HSV-2. HSV-2 infections are the leading cause of genital herpes and the incidence of HSV-2 infection has increased substantially during the past 30 years. It is estimated that in the USA, for example, from 40 to 60 million people are HSV-2 infected, with an incidence of 1 -2 million infections and 600,000-800,000 clinical cases per year. Prevalence in the 30-40 year old population is about 30%. Overall prevalence is higher in women than men, especially among the younger population. Moreover, there is increasing evidence that HSV-2 infections contribute to the spread of HIV. [0004] Herpes simplex viruses establish lifelong latent infections within sensory ganglia and therefore recurrent infections are common. Reactivation of virus, i.e. release from latency, occurs periodically over the lifetime of the individual and may result in recurrent infection but always involves virus shedding.
[0005] The structure of herpes viruses consists of a relatively large double-stranded, linear DNA genome encased within an icosahedral protein cage called the capsid, which is wrapped in a lipid bilayer called the envelope. The HSV envelope harbors 12 surface proteins and glycoproteins. To deliver the capsid containing the double-stranded DNA genome into the host cell, the virus fuses its envelope with the host's cellular membrane, either at the cell surface or within an endocytic vesicle, depending on the cell type. Among the envelope glycoproteins, gC, gB, gD, gH, and gL participate in viral-cell binding and entry.
[0006] HSV remains a significant human pathogen in spite of effective anti-viral therapy. Anti-viral drugs, such as acyclovir, used to treat HSV infections and recurrences, target DNA replication. However, these drugs are administered only when clinical signs appear, but virus shedding occurs even in the absence of clinical signs and is a major source of spread in the human population. Thus, there is an urgent need for an effective HSV vaccine both for prophylactic use and therapeutic use.
SUMMARY OF THE INVENTION
[0007] The present invention provides isolated HSV glycoprotein subunits acting as antigens, wherein the subunits are isolated or recombinantly produced, and utilized as compositions for the vaccination of mammalian subjects. In some embodiments, the glycoprotein subunit can be encoded by an isolated nucleic acid molecule.
[0008] The present invention includes compositions and methods of using such compositions to provide a therapeutic or prophylactic effect against HSV-1 and HSV-2. More particularly, the present invention relates to methods and compositions for HSV vaccines. The present invention provides for the use of three or more HSV glycoproteins, optionally, in combination with an adjuvant to vaccinate a mammalian subject against HSV viral strains. In particular embodiments, the composition includes glycoprotein B2 (gB2) and glycoprotein H2 (gH2) complexed with glycoprotein L2 (gL2). In some aspects, the composition further contains glycoprotein D2 (gD2).
[0009] In a first embodiment of the invention, there are provided compositions of isolated Herpes Simplex Virus (HSV) antigens including an isolated glycoprotein B (gB) or a fragment thereof, and an isolated glycoprotein H (gH) or a fragment thereof in a complex with glycoprotein L (gL) of a fragment thereof. In certain embodiments, the composition further includes an isolated glycoprotein D (gD) or fragment thereof. In particular embodiments, the glycoproteins are from HSV Type 2 (HSV-2). In one aspect, the glycoprotein B is glycoprotein B2 (gB2), the glycoprotein H is glycoprotein H2 (gH2), the glycoprotein L is glycoprotein L2 (gL2), and the glycoprotein D is glycoprotein D2 (gD2). In another aspect, the glycoprotein B is glycoprotein Bl (gBl), the glycoprotein H is glycoprotein HI (gHl), the glycoprotein L is glycoprotein LI (gLl), and the glycoprotein D is glycoprotein Dl (gDl).
[0010] In some embodiments of the present compositions and methods, the glycoprotein fragment is the ectodomain of the full length glycoprotein. For example, in particular embodiments, the fragment of gB includes amino acid residues 31-726 or amino acid residues 31-727 of gB2, and the fragment of gH includes amino acid residues 21-802 of gH2. In other embodiments, the fragment of gB includes amino acid residues 31-726 or amino acid residues 31-727 of gB2, the fragment of gH includes amino acid residues 21-802 of gH2, and the fragment of gD comprises amino acid residues 1-285 of gD2.
[0011] In particular embodiments there are provided, compositions of isolated herpes simplex virus (HSV) antigens including an isolated polypeptide containing amino acid residues 31-726 of glycoprotein B2 (gB2), and an isolated polypeptide containing amino acid residues 21-802 of glycoprotein H2 (gH2) in a complex with glycoprotein L2 (gL2). In one aspect, the composition further contains an isolated polypeptide containing amino acid residues 1-285 of glycoprotein D2 (gD2).
[0012] In preferred embodiments, the compositions of glycoproteins further contain an adjuvant. In one aspect, the adjuvant is a cationic liposome DNA complex (CLDC). In certain embodiments, the CLDC comprises a cationic liposome delivery vehicle and an isolated nucleic acid molecule. The isolated nucleic acid molecule is selected from the group consisting of i) an isolated nucleic acid molecule that is not operatively linked to a transcription control sequence, ii) an isolated bacterially-derived nucleic acid vector without a gene insert, iii) an isolated nucleic acid molecule comprising a non-coding nucleic acid sequence; iv) an isolated recombinant nucleic acid molecule encoding an immunogen operatively linked to a transcription control sequence, and iv) an oligonucleotide comprising a CpG motif. In particular embodiments, the composition contains HSV antigens including an isolated glycoprotein B (gB) or a fragment thereof, and an isolated glycoprotein H (gH) or a fragment thereof in a complex with glycoprotein L (gL) of a fragment thereof, and optionally, an isolated glycoprotein D (gD) or fragment thereof, wherein the composition is in CLDC adjuvant and the CLDC includes nucleic acid molecules encoding one or more of the antigens.
[0013] In other embodiments of the invention there are provided nucleic acid molecules encoding one or more of the antigens of the invention compositions or methods. In one aspect, the nucleic acid molecule encodes gH and gL. In another aspect the nucleic acid molecule encodes gH, gL, and gB. In still another aspect, the nucleic acid molecule encodes gH, gL, gB, and gD.
[0014] In other embodiments, there are provided compositions including an isolated nucleic acid molecule encoding a polypeptide containing amino acid residues 31-726 of glycoprotein B2 (gB2), and an isolated nucleic acid molecule encoding a polypeptide containing amino acid residues 21-802 of glycoprotein H2 (gH2), and an isolated nucleic acid molecule encoding glycoprotein L2 (gL2). In one aspect, the nucleic acids are complexed with a cationic liposome delivery vehicle to form a cationic liposome DNA complex
(CLDC).
[0015] In certain embodiments, the compositions are for use in inducing an immune response against HSV in a mammalian subject. In some aspects, the use is for vaccinating a mammalian subject against HSV. In one aspect, the use is for vaccinating a mammalian subject against HSV-2. In some embodiments, the composition is effective to treat an HSV-2 infection. In some embodiments, the mammalian subject is a human. In one aspect, the subject has been diagnosed with an HSV-2 infection.
[0016] Compositions contemplated for vaccinating a mammalian subject against
HSV-1 and -2 preferably include one or more antigenic herpes subunit proteins with a CLDC. Compositions contemplated for vaccinating a mammalian subject against HSV-1 or -2, may feature a full length, truncated, or mutated form of gB2, gH2/gL2 (a single subunit or multivalent subunits), and optionally, gD2. In particular aspects, the composition includes an adjuvant.
[0017] Additional embodiments of the featured compositions may include liposome delivery vehicles comprising lipids selected from the group consisting of multilamellar vesicle lipids and extruded lipids. Additional liposome delivery vehicle embodiments may include pairs of lipids selected from the group consisting of DOTMA and cholesterol;
DOTAP and cholesterol; DOTIM and cholesterol; and DDAB and cholesterol.
[0018] Additional embodiments feature methods of vaccinating a mammalian subject against herpes virus by administering one of the compositions embodied in the present invention.
[0019] In another embodiment of the invention, there are provided methods for vaccinating a mammalian subject against HSV-2 by concurrently or sequentially
administering to the subject an effective amount of an isolated polypeptide containing amino acid residues 31-726 of glycoprotein B2 (gB2) in an adjuvant, and an isolated polypeptide containing amino acid residues 21-802 of glycoprotein H2 (gH2) in a complex with glycoprotein L2 (gL2), wherein the complex is in an adjuvant. In particular embodiments, the method further includes concurrently or sequentially administering an isolated polypeptide comprising amino acid residues 1-285 of glycoprotein D2 (gD2) in an adjuvant. In one aspect, the adjuvant is a cationic liposome DNA complex (CLDC).
[0020] In still another embodiment of the invention, there are provided methods for vaccinating a mammalian subject against HSV-2 comprising concurrently or sequentially administering to the subject an effective amount of an isolated nucleic acid molecule encoding a polypeptide comprising amino acid residues 31 -726 of glycoprotein B2 (gB2) complexed with a cationic liposome delivery vehicle, and an isolated nucleic acid molecule encoding a polypeptide comprising amino acid residues 21-802 of glycoprotein H2 (gH2) in combination with an isolated nucleic acid molecule encoding a glycoprotein L2 (gL2), wherein the combination is complexed with a cationic liposome delivery vehicle. In particular embodiments, the method further includes concurrently or sequentially
administering an isolated nucleic acid molecule encoding a polypeptide comprising amino acid residues 1-285 of glycoprotein D2 (gD2) complexed with a cationic liposome delivery vehicle.
BRIEF DESCRIPTION OF THE DRAWINGS
[0021] The accompanying drawings, which are incorporated in and form a part of the specification, illustrates embodiments of the present invention, and together with the description serve to explain the principles of the invention.
[0022] Figure 1 shows a plot of the percentage of mice, which were vaccinated as indicated, exhibiting HSV symptoms over a period of 21 days post-HSV-2 infection. Clinical outcome through day 21 in mice challenged with HSV-2 is shown. 60 mice (i.e., 12 mice/group with five groups) were either vaccinated according to the following groups:
Group 1) no HVAC/no adjuvant; Group 2) whole detergent-inactivated HSV-2 lysate (HVAC) alone; Group 3) HVAC + CLDC; Group 4) HVAC +MPL (3-deacylated
monophosphoryl lipid A); or Group 5) CLDC alone.
[0023] Figure 2 provides a plot of survival to day 21 following intravaginal HSV-2 challenge of the mice analyzed in Figure 1. [0024] Figure 3 provides a plot of survival to day 30 following intravaginal HSV-2 challenge, of mice (12 mice/group with four groups), which were vaccinated as follows: Group 1) no HVAC/no adjuvant; Group 2) HVAC alone; Group 3) HVAC + CLDC; and Group 4: HVAC + MPL.
[0025] Figure 4 shows plots of the antibody responses to gD2 vaccines with CLDC or
MPL/alum (aluminium hydroxide). The mice were divided into five groups: Group 1) no HVAC/no adjuvant; Group 2) gD2 alone; Group 3) gD2 + CLDC; Group 4) gD2 +
MPL/alum; and Group 5) HVAC + CLDC.
[0026] Figure 5 provides a plot of survival to 30 days following intravaginal HSV-2 challenge of mice, vaccinated according to the following groups: Group 1) was no gD2/no adjuvant; Group 2) gD2 alone; Group 3) gD2 + CLDC; Group 4) gD2 + MPL/alum; and Group 5) HVAC + CLDC.
[0027] Figure 6 shows a plot of the interferon-gamma (IFN- γ) response of splenocytes to gD2 vaccination. The mice were vaccinated with gD2 alone, gD2 + CLDC, or diluent.
[0028] Figure 7 provides the clinical outcome of primary lesions in guinea pigs following intravaginal HSV-2 challenge. Sixty guinea pigs were randomized into five groups (12 guinea pigs/group): Group 1) no vaccine; Group 2) CLDC alone; Group 3) gD2 alone; Group 4) gD2 + CLDC; and Group 5) gD2 + MPL/alum.
[0029] Figure 8 shows a plot of the primary vaginal virus replication following HS V -
2 challenge. Vaginal swabs were collected from the same five groups of guinea pigs as those in Figure 7. Virus titers were determined by plaque assay. [0030] Figure 9 shows a plot of the effects of vaccination on subsequent recurrent disease from 3 to 9 weeks after challenge of the five groups of guinea pigs from Figure 8.
[0031] Figure 10 provides a plot of the percentage of guinea pigs in each of the indicated vaccination group with subsequent recurrent lesions.
[0032] Figure 11 shows a plot of the mean number of days with recurrent lesions for each of the indicated vaccination groups of guinea pigs.
[0033] Figure 12 provides a plot of the number of animals that shed virus is each of the indicated vaccination groups. Mean shedding days are mean number of days in which viral DNA was detected from day 21 to day 63 following HSV-2 challenge.
[0034] Figure 13 shows a plot of the latent viral DNA in the dorsal root ganglia
(DRG) after the indicated rounds of PCR for five groups of guinea pigs (12 guinea pigs/group): Group 1) no vaccine; Group 2) gD2-FL + CLDC; Group 3) gD2T + CLDC; Group 4) gD2T +gB2T+gH2/gL2+CLDC and Group 5) gB2 +gH2/gL2+CLDC
[0035] Figure 14 shows a plot of the recurrent mean cumulative lesion scores for the indicated vaccination groups.
[0036] Figure 15 shows a plot of the mean cumulative lesion days for the indicated vaccination groups.
[0037] Figure 16 shows plots of mouse percent survival and disease progression scores in groups of mice vaccinated with the indicated antigen(s) and with adjuvant (plots on the right side) or without adjuvant (plots on the left side). Animals were monitored and scored for survival and clinical disease signs after intravaginal challenge with HSV-2 virus. [0038] Figure 17 shows plots of mouse vaginal viral titers of groups of mice vaccinated with the indicated antigen(s) and with adjuvant (plot on the right side) or without adjuvant (plots on the left side). Viral titer was assayed by culture of vaginal swabs for the presence of HSV-2 virus at 2, 4, and 6 days post viral challenge.
[0039] Figure 18 shows a plot of mouse antigen specific antibody titers from groups of mice vaccinated with the indicated combinations of HSV-2 antigen and adjuvant.
Antibody titers were assayed for antigen specific binding antibody. Average logECso, the log dilution at the midpoint of the binding curve, are shown for each group against gD (black bars), gB (grey bars), and gH/gL (white bars) antigens. Anti-gD antibodies cross reacted with gB but not vice versa.
[0040] Figure 19 shows the results of assays to detect T-cell specific responses to the vaccines in mice. The graphs represent the frequencies of IFNy (bottom two plots), TNFa (middle two plots), and IL-2 (top two plots) cytokine producing CD4+ (left side plots) and CD8+ (right side plots) induced for each vaccination group.
[0041] Figure 20 shows a plot of antigen specific antibody titers in non-human primates. Groups of monkeys vaccinated with the indicated combinations of HSV-2 antigen and adjuvant were assayed for antigen specific binding antibody. Average logEC50, the log dilution at the midpoint of the binding curve, are shown for each group against gD (black bars), gB (grey bars), and gH/gL (white bars) antigens. Anti-gD antibodies cross react with gB and gH/gL.
[0042] Figure 21 shows the results of assays to detect HSV2 T cell specific responses in non-human primate. The graphs represent the frequencies of IFNy, TNFa, and IL-2 cytokine producing CD4+ (bottom plot) and CD8+ (top plot) induced for each vaccination group.
[0043] Figure 22 sets forth the amino acid sequence (SEQ ID NO:l) and nucleic acid sequence (SEQ ID NO:2) of an exemplary HSV-2 glycoprotein D (gD2; GenBank Accession No. ABU45462.1 ; GenBank ID Nos. GI: 156072227 and GI: 156072226).
[0044] Figure 23 sets forth the amino acid sequence (SEQ ID NO:3) and nucleic acid sequence (SEQ ID NO:3) of an exemplary HSV-2 glycoprotein B (gB2; GenBank ID Nos. GL295848592 and GI:295848591).
[0045] Figure 24 sets forth the amino acid sequence (SEQ ID NO: 5) and nucleic acid sequence (SEQ ID NO:6) of an exemplary HSV-2 glycoprotein H (gH2; GenBank ID No. GI:82013506).
[0046] Figure 25 sets forth the amino acid sequence (SEQ ID NO:7) and nucleic acid sequence (SEQ ID NO:8) of an exemplary HSV-2 glycoprotein L (gL2; GenBank ID No. GI: 136776).
[0047] Figure 26 sets forth the amino acid sequence (SEQ ID NO: 9) and nucleic acid sequence (SEQ ID NO: 10) of an exemplary HSV-1 glycoprotein D (gDl ; GenBank ID Nos. GI: 330101 and GI: 330100).
[0048] Figure 27 sets forth the amino acid sequence (SEQ ID NO: 11) and nucleic acid sequence (SEQ ID NO: 12) of an exemplary HSV-1 glycoprotein B (gBl; GenBank ID Nos. GI: 122831535 and GI: 122831534). [0049] Figure 28 sets forth the amino acid sequence (SEQ ID NO: 13) and nucleic acid sequence (SEQ ID NO: 14) of an exemplary HSV-1 glycoprotein H (gHl ; GenBank ID Nos. GI: 290766017 and 290766003).
[0050] Figure 29 sets forth the amino acid sequence (SEQ ID NO: 15) and nucleic acid sequence (SEQ ID NO: 16) of an exemplary HSV-1 glycoprotein L (gLl ; GenBank ID Nos. GI: 32344856 and 32344855).
DETAILED DESCRIPTION OF THE INVENTION
[0051] HSV glycoproteins function in virus entry, spread, immune evasion and inhibition of apoptosis. These glycoproteins include gD, the receptor binding protein, gB, and the gH/gL complex, which contribute to the release of the capsid inside the cell to begin replication. Other glycoproteins, such as gC and gE, also are involved in the pathway of virus invasion and infection. The virus cascade of events consists of a series of coordinated steps and conformational changes.
[0052] Both the humoral and cell-mediated immune responses are important for controlling HSV infection. Although both CD4+ and CD8+ T cells play an important role in protection from HSV infection in humans and in murine models, the construction of an effective prophylactic or therapeutic vaccine against HSV is still needed. The present invention identifies particular immunogenic glycoprotein antigens, with or without an adjuvant to enhance the immune response. Herpes simplex viruses.
[0053] Billions of people worldwide are infected with either Herpes simplex virus type 1 (HSV-1) or Herpes simplex virus type 2 (HSV-2). HSV-1 and HSV-2 are two members of the HSV family of oc-herpesviruses, which establish lifelong latent infection in sensory neurons and lead to chronic herpes disease. HSV-1 infection causes facial/ocular disease, while HSV-2 is the leading cause of genital herpes, although both viruses can be found at oral and genital sites. Indeed, the incidence of HSV-1 genital disease is increasing and approximates that of HSV-2 in certain countries. HSV-1 and HSV-2 encode
approximately 80 genes, are structurally similar, and share approximately 80% genome sequence homology.
[0054] Although many, if not most, HSV-2 infections are asymptomatic or unrecognized, symptomatic primary genital HSV infection is characterized by vesicular and ulcerative skin lesions, which can result in neurologic and urologic complications. During initial infection, a long term persistent or latent infection is established in ganglion neurons, which can reactivate and cause recurrent genital disease or asymptomatic viral shedding. Recurrent herpes infection is a chronic, intermittent disease characterized by both
symptomatic and asymptomatic periods of viral replication in the epithelial cells at mucosal sites or other peripheral sites. (Koelle DM et al., J Immunol. 166:4049, 2001). Both symptomatic and asymptomatic herpes infection can lead to transmission to seronegative individuals. Indeed, most transmission occurs during periods of unrecognized shedding. One of the most serious complications of genital herpes occurs when the virus is transmitted from mother to neonate. Infection of the neonate causes significant morbidity and mortality, even with proper antiviral therapy (Sacks SL et al., Antiviral Res. 63S1 :S27, 2004). Genital herpes infection also increases the risk of acquiring HIV infection and increases shedding of HIV in genital lesions (Posovad CM et al., Proc Natl Acad Sci USA. 94: 10289, 1997).
[0055] HSV-2 infection induces both humoral and T-cell mediated immunity;
however, the mechanisms that contribute to long term control of genital herpes are not understood. Studies from animal models of HSV infection and human studies indicate that high levels of neutralizing antibodies and innate immunity (natural killer (NK) cells, interferon, and macrophages) contribute to protection from HSV infection but the major determinants of HSV protection are both CD4+ and CD8+ T cells (Ahmad A et al., J Virol. 74(16): 7196-7203, 2000; Aurelian L et al., J Gen Virol. 68:2831, 1987; Milligan GN et al., J Immunol. 160(12):6093, 1998.; Koelle et al, 1998). Clearance of virus from recurrent lesions is more closely correlated to T cell immunity. Thus, when a recurrent lesion occurs, mononuclear cells, primarily CD4+ T cells, infiltrate the lesion as early as two days after formation, followed by an influx of CD8+ T cells at later times (Cunningham AL et al., J Clin Invest. 75:226, 1985). Although both HSV-specific CD4+ and CD8+ T cell responses are detected, clearance of HSV-2 from lesions correlates with a CD8+ cytotoxic T lymphocyte (CTL) response (Koelle et al., J Clin Invest. 101 : 1500, 1998). To counteract the host immune response, HSV has developed a number of immune evasion strategies, such as disruption of antigen peptide processing and presentation by MHC Class I molecules (Jugovic P et al., J Virol. 72:5076, 1998; Tomazin R et al., J Virol. 72(3):2560, 1998), interference with complement activation and antibody binding (Friedman HM et al., J Immunol. 165:4528, 2000; Nagashunmugam T et al., J Virol. 72:5351, 1998), or inhibition of dendritic cell maturation, which reduces efficient antigen presentation and CTL priming (Salio M et al., Eur J Immunol. 29:3245, 1999; Pollara G et al., J Infect Dis. 187: 1513, 2003). IFNy, a Thl cytokine, is expressed in lesions by T cells (predominantly CD4+), and partially restores MHC I expression and also stimulates MHC II expression, thus contributing to T cell- mediated immune control of HSV replication (Cunningham AL et al., J Clin Investig. 83:490, 1989; Kokuba H et al, J Investig Dermatol. 113:808, 1999; Mikloska Z et al., J Gen Virol. 79:353, 1998). Such mechanisms provide HSV with strategies to counteract the immune system and establish a lifelong persistent infection in the host.
[0056] HSV has a lipid envelope, bearing about 12 surface proteins and
glycoproteins. To deliver the capsid containing the double-stranded DNA genome into the host cell, HSV must fuse its envelope with a cellular membrane, either at the cell surface or within an endocytic vesicle, depending on the cell type. Among the envelope glycoproteins, gC, gB, gD, gH, and gL, appear to participate in viral cell binding and entry.
[0057] All herpesviruses encode genes for gB, gH, and gL, and these three proteins constitute their core fusion machinery. HSV attaches to cells by binding to heparan sulfate proteoglycan (HSPG) via gC, and by binding of gD to one of three cellular receptors: nectin- 1, herpesvirus entry mediator (HVEM), or a modified heparan sulfate. Structural and biochemical studies have shown that gD undergoes conformational changes to permit receptor binding and to potentiate fusion.
[0058] Binding of gD to its receptor nectin-1 triggers an interaction between gB and gH/gL that is on the pathway to fusion. Based on what happens to other viral fusion proteins upon activation of the triggering signal (either receptor binding or lowered pH or both), it is likely that receptor binding by gD (and possibly pH as well) causes gB and/or gH/gL to interact and to undergo conformational changes that bring the viral and cell membranes together to facilitate fusion. [0059] Recombinant Herpes Simples Virus glycoprotein D, glycoprotein B, glycoprotein H, and glycoprotein L of both Types 1 and 2 are provided, including fragments, precursors and analogs, as well as DNA sequences encoding for the precursors and the mature amino acid sequence and fragments thereof.
[0060] As used herein, the term "precursor," when used in reference to a protein refers to an amino acid sequence that includes an N-terminal signal peptide and the mature protein. Accordingly, the terms "mature protein" or "full length protein" refer to the amino acid sequence including all domains of the native sequence except the signal peptide.
[0061] As used herein, the term "ectodomain" refers to the portion of an integral membrane protein that extends into the extracellular space (i.e., the space outside the cell). The ectodomain generally does not include the signal peptide. The ectodomain of an integral membrane protein can be readily identified by the skilled artisan using sequence analysis tools known in the art. In preferred embodiments of the present compositions and methods, fragments corresponding to the ectodomain of each glycoprotein are used. The ectodomains of exemplary gD, gB, and gH proteins from HSV-1 (termed gDl, gBl, and gHl) and HSV-2 (termed gD2, gB2, and gH2) are described below. Full-length or truncated forms of the ectodomain may be used in the present compositions and methods.
[0062] Figures 22-29 provide the amino acid and encoding nucleotide sequences of exemplary gD, gB, gH, and gL proteins from HSV-1 and HSV-2. The nucleotide sequences encoding a glycoprotein may be varied by methods known to the skilled artisan. For example, a nucleotide sequence encoding an entire precursor or mature glycoprotein polypeptide, or a portion thereof may be joined in-frame to a nucleotide sequence encoding one or more different precursor or mature glycoprotein polypeptides, or a portions thereof. In this way, a single nucleic acid molecule may be generated that encodes two or more glycoprotein polypeptides or portions thereof. In another example, a nucleotide sequence encoding a portion of a precursor or mature glycoprotein polypeptide may be joined in-frame with another portion of the same glycoprotein to generate a nucleic acid molecule encoding a glycoprotein having a deletion. In preferred embodiments, the transmembrane domain or anchor is deleted from glycoproteins that are membrane proteins.
[0063] In addition, particular codons may be modified so as the encode the same amino acids, but provide more efficient expression in accordance with the nature of the host. For example, the codons may be modified in accordance with the frequency of occurrence of a particular codon in one or more proteins or groups of proteins, e.g., glycolytic proteins, which contribute to a high proportion of the total proteins of a particular host, e.g., human. In some instances one or more codons may be modified to code for a different amino acid, substituting one amino acid for another amino acid, where the effect of the change is not detrimental to the immunogenicity of the protein or to other biological factors of interest. Such changes may be made to enhance the immunogenicity of the polypeptide, facilitate conjugating the polypeptide to a carrier protein, or the like. It may be desirable in some instances to add amino acids to the N-terminus or C-terminus, where such additional amino acids may provide-for a desired result. This can be readily achieved by providing for additional codons at the 5'- or 3'- termini of the sequence encoding the mature glycoprotein or its precursor.
HSV glycoprotein D (gD).
[0064] gD is a type I integral membrane protein with an ectodomain of 316 residues organized around a core immunoglobulin (Ig) V-fold with N- and C-terminal extensions that wrap around the core. Most receptor binding activities and conformational changes involve the N-and C-terminal extensions.
[0065] In the unbound form, the C-terminus folds back around the Ig fold and is anchored near the N-terminus. Nectin-1 binds to the gD core at a position distinct from where HVEM binds. However, the gD C-terminus normally obscures the binding sites for both nectin-1 and HVEM and necessitates displacement of this region. Furthermore, this conformational change to gD and/or the residues of gD that are exposed by receptor binding likely activate the fusion machinery. Triggering of fusion by gD/receptor binding would occur only when the virus is close to a membrane, thereby preventing premature activation of the fusion machinery. The gD protein also includes proline-rich region (a.a. 260-285), termed the profusion domain (PFD) that is important for gD function after receptor binding. A portion of this segment is unresolved in all X-ray structures, suggesting it is highly flexible. It may act as a hinge that allows another part of gD to activate the fusion protein(s) or contact these proteins directly. The PFD concept agrees with structural studies regarding the ectodomain C-terminus. Analogous proline-rich regions connect functional domains of other viral envelope proteins. Alternatively, the conformational change that takes place upon receptor binding may expose key residues of the gD Ig core, normally hidden, that are directly involved in triggering downstream events involving gB and gH/gL.
[0066] HSV-2 glycoprotein D contains 393 amino acid residues. An exemplary gD2 amino acid sequence is set forth in GenBank ID No. GI: 156072227, which is provided in Figure 22 as SEQ ID NO: l . The signal sequence of gD2 encompasses residues -25 to -1 of the sequence set forth in Figure 22, and the ectodomain of gD2 encompasses approximately residues 1-316, with a soluble gD2 ectodomain encompassing residues 1-306. The skilled artisan will readily recognize that the referenced specific sequence was obtained from gD2 from a particular HSV-2 strain and that the analogous sequences from another HSV-2 strain may vary. Accordingly, in some embodiments of the present compositions and methods, the gD2 antigen may be 80% identical to the sequence set forth in SEQ ID NO: 1. In other embodiments, gD2 antigen may be 85%, 90%, 95%, 96%, 97%, 98%, or even 99% identical to the sequence set forth in SEQ ID NO: 1. The skilled artisan can readily identify and confirm the immunogenicity of these sequences using methods known in the art and provided herein. In some embodiments of the present compositions and methods, a full length gD2 protein is used. In some aspects, the ectodomain of gD2 is used. In other embodiments, a truncated, mutated, or chimeric form of gD2 is used. For example, truncated form gD- 2t(285) contains residues 1-285 and is a preferred aspect of the invention. Other truncated forms include those with deletions between residues 260-306; preferably between 275-285 resulting in truncated forms such as amino acid residues 1-260 and 1-275. (Krummenacher et al. 2005 EMBO Journal 24:4144-4153.)
[0067] HSV-1 glycoprotein D contains 394 amino acid residues. An exemplary gDl amino acid sequence is set forth in GenBank ID No. GI: 330101, which is provided in Figure 26 as SEQ ID NO:9. The signal sequence of gDl encompasses residues -25 to -1 of the sequence set forth in Figure 26 and the ectodomain of gDl encompasses residues 1-316. The skilled artisan will readily recognize that the referenced specific sequence was obtained from gDl from a particular HSV-1 strain and that the analogous sequences from another HSV-1 strain may vary. Accordingly, in some embodiments of the present compositions and methods, the gDl antigen may be 80% identical to the sequence set forth in SEQ ID NO:9. In other embodiments, gD2 antigen may be 85%, 90%, 95%, 96%, 97%, 98%, or even 99% identical to the sequence set forth in SEQ ID NO:9. The skilled artisan can readily identify and confirm the immunogenicity of these sequences using methods known in the art and provided herein. In some embodiments of the present compositions and methods, a full length gDl protein is used. In some aspects, the ectodomain of gDl is used. In other embodiments, a truncated, mutated, or chimeric form of gDl is used. For example, truncated form gD-lt(285) contains residues 1-285 and is a preferred aspect of the invention. Other truncated forms include those with deletions between residues 260-306; preferably between 275-285 resulting in truncated forms such as amino acid residues 1-260 and 1-275.
[0068] Various aspects of the gDl and gD2 glycoprotein are found in US Patents
4,762, 708; 5,149,660; 5,654,174; and 5,814,486.
HSV glycoprotein B (gB).
[0069] The gB glycoprotein is an elongated trimer with a long central alpha-helix in each protomer. Each protomer contains five distinct structural domains. Domains I and II have features that resemble those of pleckstrin homology (PH) domains. Remarkably, much of the architecture of gB is similar to that of the post-fusion form of VSV G, implying that gB is a viral fusogen and that the solved structure of gB may be a post-fusion form. The alpha- helical coiled-coil core relates gB to class I viral membrane fusion glycoproteins. However, both gB and VSV G have two extended beta hairpins with hydrophobic tips (domain I of gB and domain IV of VSVG) that are homologous to fusion loops of class II fusion proteins. Members of both classes accomplish fusion through a large-scale conformational change (fold-back) triggered by a signal from a receptor-binding component. It is likely that gB also undergoes a fold-back type of change.
[0070] HSV-2 glycoprotein B contains 901 amino acid residues. The gB protein may be divided into four domains beginning at the N-terminus: a signal sequence, followed by an ectodomain, a transmembrane domain or "anchor," and a cytoplasmic domain. An exemplary gB2 amino acid sequence is set forth in GenBank ID No. GI:295848592, which is provided in Figure 23 as SEQ ID NO:3. The ectodomain of gB2 encompasses approximately residues 31 -726. The skilled artisan will readily recognize that the referenced specific sequence was obtained from gB2 from a particular HSV-2 strain and that the analogous sequences from another HSV-2 strain may vary. Accordingly, in some embodiments of the present compositions and methods, the gB2 antigen may be 80% identical to the sequence set forth in SEQ ID NO:3. In other embodiments, gB2 antigen may be 85%, 90%, 95%, 96%, 97%, 98%, or even 99% identical to the sequence set forth in SEQ ID NO:3. The skilled artisan can readily identify and confirm the immunogenicity of these sequences using methods known in the art and provided herein. In some embodiments of the present compositions and methods, a full length gB2 protein is used. In some aspects, the ectodomain of gB2 is used. In other embodiments, a truncated, mutated, or chimeric form of gB2 is used. For example, a truncated form gB2 containing residues 31-726 and is a preferred aspect of the invention. In another preferred aspect, a truncated form gB2 containing residues 31-727 is used in the disclosed compositions and methods.
[0071] HSV-1 glycoprotein B may contain 899-904 amino acid residues, depending on the strain from which it is obtained. The gB protein may be divided into four domains beginning at the N-terminus: a signal sequence, followed by an ectodomain, a transmembrane domain or "anchor," and a cytoplasmic domain. An exemplary gBl amino acid sequence is set forth in GenBank ID No. GI: 122831535, which is provided in Figure 27 as SEQ ID NO: 1 1. The ectodomain of gBl encompasses approximately residues 34-727. The skilled artisan will readily recognize that the referenced specific sequence was obtained from gBl from a particular HSV-1 strain and that the analogous sequences from another HSV-1 strain may vary. Accordingly, in some embodiments of the present compositions and methods, the gBl antigen may be 80% identical to the sequence set forth in SEQ ID NO: 11. In other embodiments, gBl antigen may be 85%, 90%, 95%, 96%, 97%, 98%, or even 99% identical to the sequence set forth in SEQ ID NO: 11. The skilled artisan can readily identify and confirm the immunogenicity of these sequences using methods known in the art and provided herein. In some embodiments of the present compositions and methods, a full length gBl protein is used. In some aspects, the ectodomain of gBl is used. In other embodiments, a truncated, mutated, or chimeric form of gBl is used. For example, a truncated form gBl containing residues 31-727 and is a preferred aspect of the invention. In another preferred aspect, a truncated form gBl containing residues 31-728 is used in the disclosed
compositions and methods.
[0072] Various gB glycoproteins also are described in US Patent Nos. 5,244,792;
5,648,079; and 5,759,814.
HSV glycoprotein H/L (gH/gL).
[0073] Glycoproteins gH and gL form a non-covalently linked heterodimer that is associated with HSV entry and cell fusion. The proper processing and transport of gH requires it to be co-expressed with gL as a heterodimer. For example, it has been shown that when HSV- 1 gH is expressed in the absence of gL, it is retained in the endoplasmic reticulum in an antigenically and structurally immature form. Therefore, gL is a chaperone for gH transport, but it may play an additional role, as some anti-gL MAbs inhibit virus spread. Interestingly, HSV gL contains an N-terminal signal peptide sequence, but lacks a hydrophobic transmembrane region (TMR). (See US Patents 5,807,557; 6,156,319; and 6,541,459.) HSV-2 contains a similar complex of gH2/gL2. [0074] HSV-2 glycoprotein H is a single pass type I membrane protein and contains
838 amino acid residues. The gH protein may be divided into four domains beginning at the N-terminus: a signal sequence, followed by an ectodomain, a transmembrane domain or "anchor," and a cytoplasmic domain. An exemplary gH2 amino acid sequence is set forth in GenBank ID No. GI:82013506, which is provided in Figure 24 as SEQ ID NO:5. The full ectodomain of gH2 encompasses approximately residues 19-803. The skilled artisan will readily recognize that the referenced specific sequence was obtained from gH2 from a particular HSV-2 strain and that the analogous sequences from another HSV-2 strain may vary. Accordingly, in some embodiments of the present compositions and methods, the gH2 antigen may be 80% identical to the sequence set forth in SEQ ID NO:5. In other embodiments, gH2 antigen may be 85%, 90%, 95%, 96%, 97%, 98%, or even 99% identical to the sequence set forth in SEQ ID NO: 5. The skilled artisan can readily identify and confirm the immunogenicity of these sequences using methods known in the art and provided herein. In some embodiments of the present compositions and methods, a full length gH2 protein is used. In some aspects, the ectodomain of gH2 is used. In other embodiments, a truncated, mutated, or chimeric form of gH2 is used. For example, a truncated form gH2 containing residues 21-802 and is a preferred aspect of the invention.
[0075] HSV-1 glycoprotein H is a single pass type I membrane protein and contains
838 amino acid residues. The gH protein may be divided into four domains beginning at the N-terminus: a signal sequence, followed by an ectodomain, a transmembrane domain or "anchor," and a cytoplasmic domain. An exemplary gHl amino acid sequence is set forth in GenBank ID No. GI: 290766017, which is provided in Figure 28 as SEQ ID NO: 13. The ectodomain of gHl encompasses approximately residues 19-803. The skilled artisan will readily recognize that the referenced specific sequence was obtained from gHl from a particular HSV-1 strain and that the analogous sequences from another HSV-1 strain may vary. Accordingly, in some embodiments of the present compositions and methods, the gHl antigen may be 80% identical to the sequence set forth in SEQ ID NO: 13. In other embodiments, gHl antigen may be 85%, 90%, 95%, 96%, 97%, 98%, or even 99% identical to the sequence set forth in SEQ ID NO: 13. The skilled artisan can readily identify and confirm the immunogenicity of these sequences using methods known in the art and provided herein. In some embodiments of the present compositions and methods, a full length gHl protein is used. In some aspects, the ectodomain of gHl is used. In other embodiments, a truncated, mutated, or chimeric form of gHl is used. For example, a truncated form gHl containing residues 21-802 and is a preferred aspect of the invention. The skilled artisan will recognize that the positions of the recited truncation may differ slightly in glycoproteins from different HSV strains, and can readily identify the analogous sequences using well-known sequence analysis tools.
[0076] HSV-2 glycoprotein L is viral envelope protein and contains 224 amino acid residues. An exemplary gL2 amino acid sequence is set forth in GenBank ID No.
GI: 136776, which is provided in Figure 25 as SEQ ID NO:7. The gL protein contains a signal sequence, followed by the envelope glycoprotein L, which encompasses approximately residues 17-224. The skilled artisan will readily recognize that the referenced specific sequence was obtained from gL2 from a particular HSV-2 strain and that the analogous sequences from another HSV-2 strain may vary. Accordingly, in some embodiments of the present compositions and methods, the gL2 antigen may be 80% identical to the sequence set forth in SEQ ID NO:7. In other embodiments, gL2 antigen may be 85%, 90%, 95%, 96%, 97%, 98%, or even 99% identical to the sequence set forth in SEQ ID NO:7. The skilled artisan can readily identify and confirm the immunogenicity of these sequences using methods known in the art and provided herein. In some embodiments of the present compositions and methods, a full length gL2 protein is used. In some aspects, the glycoprotein without the signal sequence is used. In other embodiments, a truncated, mutated, or chimeric form of gL2 is used. For example, a truncated form gL2 containing residues 17-224 and is a preferred aspect of the invention.
[0077] HSV-1 glycoprotein L is viral envelope protein and contains 224 amino acid residues. An exemplary gLl amino acid sequence is set forth in GenBank ID No. GI:
32344856, which is provided in Figure 29 as SEQ ID NO: 15. The gL protein contains a signal sequence, followed by the envelope glycoprotein L, which encompasses approximately residues 17-224. The skilled artisan will readily recognize that the referenced specific sequence was obtained from gLl from a particular HSV-1 strain and that the analogous sequences from another HSV-1 strain may vary. Accordingly, in some embodiments of the present compositions and methods, the gLl antigen may be 80% identical to the sequence set forth in SEQ ID NO: 15. In other embodiments, gLl antigen may be 85%, 90%, 95%, 96%, 97%, 98%, or even 99% identical to the sequence set forth in SEQ ID NO: 15. The skilled artisan can readily identify and confirm the immunogenicity of these sequences using methods known in the art and provided herein. In some embodiments of the present compositions and methods, a full length gLl protein is used. In some aspects, the glycoprotein without the signal sequence is used. In other embodiments, a truncated, mutated, or chimeric form of gLl is used. For example, a truncated form gLl containing residues 17-224 and is a preferred aspect of the invention. Other HSV antigens
[0078] Glycoprotein gC, which is a viral envelope glycoprotein, binds to the cell surface particle: heparin sulfate; whereas gE is a membrane glycoprotein with a large ectodomain that is expressed on the virion envelope and the surface of infected cells.
Glycoprotein gA also is one of the major glycoproteins found in the envelope of the virus particle.
[0079] Additional embodiments may include the gD2, gB2, and gH2/gL2 antigens, but may also contain a human T cell HSV-2 antigen, such as ICP27, ICP4, or VP22.
HSV Vaccines
[0080] The compositions of the present invention may be used as vaccines. In one example, the vaccine includes an immunologically-effective amount of an HSV-1 or HSV-2 glycoprotein, which preferably is purified, isolated or prepared by recombinant nucleic acid methods, and an adjuvant, which is pharmaceutically or therapeutically acceptable.
[0081] A preferred composition consists essentially of isolated glycoprotein B (gB) or a fragment thereof, and an isolated glycoprotein H (gH) or a fragment thereof in a complex with glycoprotein L (gL) or a fragment thereof and adjuvant. A more preferred composition consists essentially of the ectodomain of glycoprotein B (gB), and the ectodomain of glycoprotein H (gH) or a fragment thereof in a complex with glycoprotein L (gL) and adjuvant. Another preferred composition consists essentially of an isolated glycoprotein D (gD), an isolated glycoprotein B (gB) or a fragment thereof, and an isolated glycoprotein H (gH) or a fragment thereof in a complex with glycoprotein L (gL) or a fragment thereof and adjuvant. A more preferred composition consists essentially of the ectodomain of
glycoprotein D (gD), the ectodomain of glycoprotein B (gB), and the ectodomain of glycoprotein H (gH) or a fragment thereof in a complex with glycoprotein L (gL) and adjuvant.
CLDC Adjuvant
[0082] Cationic liposome/DNA complexes (CLDC) include a cationic liposome delivery vehicle and an isolated nucleic acid molecule. In certain embodiments, the isolated nucleic acid molecule is selected from the group consisting of i) an isolated nucleic acid molecule that is not operatively linked to a transcription control sequence, ii) an isolated bacterially-derived nucleic acid vector without a gene insert, iii) an isolated nucleic acid molecule comprising a non-coding nucleic acid sequence; iv) an isolated recombinant nucleic acid molecule encoding an immunogen operatively linked to a transcription control sequence, and iv) an oligonucleotide comprising a CpG motif. In particular embodiments, the CLDC has a nucleic acid: lipid complex ratio of from about 1 : 1 to about 1 : 64. In preferred embodiments, the nucleic acid:lipid complex has a ratio of from about 1 : 10 to 1 :40. CLDC adjuvants are further described in U.S. Patent No. 6,693,086, the content of which is hereby incorporated by reference as if fully set forth herein.
[0083] Liposome delivery vehicles suitable for use in any of the compositions and methods of the present invention can include any liposomes. Particularly preferred liposomes are cationic liposomes. Other preferred liposomes include multilamellar vesicle lipids and extruded lipids, with multilamellar vesicle lipids being more preferred. Liposome
compositions can include, but are not limited to, pairs of lipids selected from DOTMA and cholesterol, DOTAP and cholesterol, DOTIM and cholesterol, and DDAB and cholesterol, with DOTAP and cholesterol being particularly preferred.
[0084] In another embodiment of the present invention, a vaccine or composition further comprises a pharmaceutically acceptable excipient. As used herein, a
"pharmaceutically acceptable excipient" refers to any substance suitable for delivering a therapeutic composition useful in the method of the present invention to a suitable in vivo or ex vivo site. Preferred pharmaceutically acceptable excipients are capable of maintaining a nucleic acid molecule and/or antigen in a form that, upon arrival of the nucleic acid molecule and/or antigen to a cell, the nucleic acid molecule and/or protein are capable of contacting and/or entering the cell and eliciting an immune response at or near the site of the cell.
[0085] Suitable excipients for use in the present invention include excipients or formularies that transport, but do not specifically target the vaccine to a cell (also referred to herein as non-targeting carriers). Examples of pharmaceutically acceptable excipients include, but are not limited to water, phosphate buffered saline, Ringer's solution, dextrose solution, serum-containing solutions, Hank's solution, other aqueous physiologically balanced solutions, oils, esters and glycols. Aqueous carriers can contain suitable auxiliary substances required to approximate the physiological conditions of the recipient, for example, by enhancing chemical stability and isotonicity. Particularly preferred excipients include non- ionic diluents, with preferred non-ionic buffer being 5%-10% sucrose or 5-10% lactose in water.
[0086] Suitable auxiliary substances include, for example, sodium acetate, sodium chloride, sodium lactate, potassium chloride, calcium chloride, and other substances used to produce phosphate buffer, Tris buffer, and bicarbonate buffer. Auxiliary substances can also include preservatives, such as thimerosal, m- or o-cresol, formalin and benzol alcohol.
Therapeutic compositions of the present invention can be sterilized by conventional methods and/or lyophilized.
Administration
[0087] The glycoprotein subunit or antigen may be administered with or without an adjuvant, such as CLDC. Moreover, one or more glycoproteins may be combined for administration with or without an adjuvant. Preferably, three or four antigens are combined, such as gB2 and gH/gL; or gB2, gD2, and gH/gL.
[0088] The single or multivalent subunit HSV vaccines may be administered to a mammal, such as a human, by methods known in the art, such as subcutaneously, intramuscularly, orally, intravenously, intradermally, intranasally or intravaginally. The antigens or subunits are formulated in a pharmaceutically acceptable excipient or carrier, which is suitable for the chosen route of administration.
[0089] Many of the above-described routes of administration, including intravenous, intraperitoneal, intradermal, and intramuscular administrations can be performed using methods standard in the art. Aerosol (inhalation) delivery can also be performed using methods standard in the art (see, for example, Stribling et al., Proc. Natl. Acad. Sci. USA 189:1 1277-11281 , 1992, which is incorporated herein by reference in its entirety). Oral delivery can be performed by complexing a composition of the present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal. Examples of such carriers, include plastic capsules or tablets, such as those known in the art. One method of local administration is by direct injection. The dose of vaccine to be used varies depending upon a number of factors, such as the age, sex, and weight of the individual as well as the chosen route of administration.
[0090] One aspect of the invention pertains to the methods for vaccinating a mammalian subject against HSV-1 or HSV-2 comprising administering to the subject an effective amount of the compositions described above. For example, a method for vaccinating a mammal, such as a human, against HSV-2 comprises concurrently or sequentially administering to the mammal an effective amount of each subunit of interest, wherein the use of an adjuvant is optional. Such adjuvants include those that induce a Th-1 immune response, including, but not limited, to, aluminium hydroxide (alum), aluminium phosphate, 3-deacylated monophosphoryl lipid A (MPL), and/or a CLDC. However, the use of CLDC is most preferred.
[0091] An effective administration protocol (i.e., administering a vaccine or therapeutic composition in an effective manner) comprises suitable dose parameters and modes of administration that result in elicitation of an immune response in a mammal that has been infected with HSV or, preferably so that the mammal is protected from infection.
Effective dose parameters can be determined using methods standard in the art for a viral infection. Such methods include, for example, determination of survival rates, side effects (i.e., toxicity) and progression or regression of disease.
[0092] A suitable single dose size is a dose that is capable of eliciting an immune response in a mammal with an HSV infection, when administered one or more times over a suitable time period. One of skill in the art can readily determine appropriate single dose sizes for systemic administration based on the size of a mammal and the route of administration. One of skill in the art can monitor the effectiveness of the immunization by measuring, for example, cytokine responses, cytotoxicity, antibody production, by enumerating antigen- specific T cells, or monitoring delayed type hypersensitivity (DTH) responses.
[0093] An appropriate single dose size for the composition based on the amount of immunogen is an amount of the vaccine that delivers from about 1 μg immunogen per individual mammal to about 50 mg immunogen per individual mammal, preferably from about 1 μg immunogen per individual mammal to about 1 mg immunogen per individual mammal, and more preferably from about 1 μg immunogen per individual mammal to about 100 μg immunogen per individual mammal, and even more preferably from about 10 μg immunogen per individual mammal to about 100 μg immunogen per individual mammal. In one embodiment, an appropriate single dose size for the composition based on the amount of immunogen is an amount of the vaccine that delivers at least about 0.1 μg immunogen per individual mammal, and more preferably at least about 1 μg immunogen per individual mammal, and m ore preferably, at least about 5 μg immunogen per individual mammal, and more preferably at least about 10 μg immunogen per individual mammal. In particular aspects the amount of immunogen delivered is from about 2-100 μg, or about 4-40 μg. One of skill in the art will appreciate that the dose amount will depend to some extent on the size of the mammal to which the composition is being administered. Approximately 1-10 doses are administered to the individual at intervals ranging from once per week to once per month to once per year.
[0094] Adjuvant, if present, is usually in the range of 10-1000 μg, preferably 250-750 μg, about 500 μg, or about 25-75 μg or about 50 μg per vaccine dose.
[0095] The examples herein are meant to exemplify the various aspects of carrying out the invention and are not intended to limit the invention in any way. EXAMPLES
Example 1
[0096] The protection of CLDC combined with a whole detergent- inactivated HSV-2 lysate (HVAC) was compared to the HVAC combined with MPL. In the initial efficacy experiment, 60 female out-bred Swiss Webster mice (12 mice/group) were divided as follows: Group 1 : no HVAC/no adjuvant; Group 2: HVAC alone; Group 3: HVAC + CLDC; Group 4: HVAC + MPL; and Group 5: CLDC alone. HSV specific antibodies were detectable after one immunization of HVAC alone or HVAC + CLDC or HVAC + MPL. Administration of CLDC enhanced the levels of detectable HSV-specific antibodies in the serum compared to mice that were immunized with HVAC + MPL (1.8-fold higher levels) or the HVAC alone (3.1 -fold higher levels) after the first dose, showing that CLDC stimulates antigen-specific antibody production. Following the second immunization, the levels of HSV specific antibodies remained highest in the HVAC + CLDC group (2195 ng/ml) compared to HVAC alone (1503 ng/ml, PO.001) or the HVAC + MPL (1758 ng/ml, P=0.012) (data not shown).
[0097] Animals were also assessed daily to monitor the development of symptomatic
HSV infection. As seen in Figure 1, 100% of the mice that received no vaccine, 83% of the mice that received CLDC alone, and 80%> of the mice that received HVAC alone, developed local disease. In contrast, only 40% of the mice that received HVAC + MPL (P=0.170 vs. vaccine alone) and 18% of the mice receiving HVAC + CLDC (P=0.009 vs. vaccine alone) developed disease. Similarly as seen in Figure 2, mice that received no vaccine, CLDC alone, or vaccine alone, were not protected from death (8-50% survival by 21 days). The HVAC + MPL protected 60% of the mice (P=0.170 vs. vaccine alone) while 100% of the mice that were vaccinated with HVAC + CLDC were protected from death (P<0.001 vs. vaccine alone and P=0.035 vs. HVAC + MPL.
[0098] To further document the antiviral nature of the vaccinations, effects on vaginal viral replication were examined. Immunization with HVAC + CLDC significantly reduced the level of infectious virus in the vagina on all four days and eliminated viral shedding by day 4 in 10 of 11 animals (data not shown). Reductions in viral shedding in the HVAC + CLDC group compared to vaccine alone was significant on all days and when compared to the HVAC + MPL group, titers were significantly less on days 2 and 3.
Example 2
[0099] A second study using 48 female out-bred Swiss Webster mice (n=12) was then performed to validate the first results and extend the observation to 30 days, as it appeared that animals in the HVAC + MPL vaccine group continued to die after day 21. In agreement with the results of the first study, mice vaccinated with the HVAC alone were more likely to develop local disease (75%) than animals vaccinated in combination with CLDC (25%, P=0.039 vs. HVAC alone). Thus, similar to the first study, HVAC + CLDC provided significantly better protection compared to HVAC + MPL (P=0.012). Significantly and as shown in Figure 3, vaccination with HVAC + CLDC protected 75% of the mice from death out to 30 days after viral challenge whereas the HVAC + MPL vaccine only protected 25% of mice from death (P=0.039), and continued to die after 21 days. These results demonstrate that the CLDC adjuvant when combined with a whole inactivated HSV-2 lysate enhances protection in the mouse model of herpes infection. Example 3
[00100] In this study, gD2 (full-length ectodomain) administered alone or with CLDC or MPL/alum was evaluated. In order to evaluate CLDC with a vaccine which more closely resembles the vaccine that previously showed some protection in clinical trials (Stanberry et al., N Engl J Med. 347(21): 1652, 2002), and is being further evaluated in a large clinical trial, the antibody and T cell response in mice immunized with gD2 (full-length ectodomain) alone, gD2 (full-length ectodomain) + CLDC, and gD2 (full-length ectodomain) + MPL/alum were analyzed. In particular, animals were immunized twice and serum obtained 3 weeks after the second immunization. The amount of anti-HSV-1 IgGl and IgG2a were measured by ELISA. As shown in Figure 4, mice receiving gD2 + CLDC, or gD2 + MPL/alum produced considerably higher levels of anti-HSV antibody than gD2 alone (P<0.01) although antibody levels induced by MPL/alum were higher compared to CLDC (P=0.05). However, vaccination with gD2 + CLDC resulted in a higher ratio of IgG2a to IgGl (0.86) compared to the gD2 + MPL/Alum group (0.39), indicating that the CLDC adjuvant is more effective than MPL/alum at promoting Thl immunity.
[00101] Vaccination with gD2 alone did not protect against disease or death, although death was delayed in mice receiving gD2 alone (mean day of death=16.0) compared to the untreated group (mean day of death=10.6, P=0.015). As seen in Figure 5, mice receiving gD2 + CLDC or gD2 + MPL/alum were significantly protected from disease (40% in each group) compared to mice receiving the gD2 antigen alone (100% with disease, (P=0.01 1). Vaccination also decreased death in the adjuvant groups from 90% in the gD2 alone group to 20% and 10%, respectively, for CLDC (P=0.006 and MPL/alum, P=0.001) (data not shown). Further, vaccination with gD2 + CLDC significantly reduced vaginal viral titers on days 1, 3, and 4 post-challenge compared to the untreated group (P<0.05) (data not shown). Similarly, mice receiving gD2 + MPL/alum had significantly lower viral titers on days 1 and 4 compared to the untreated group (P<0.05). There were no statistically significant differences in vaginal viral titers between the gD2 + CLDC and gD2 + MPL/alum groups on any day. Of interest, the virus titers in the HVAC + CLDC group were significantly less than titers to gD2 + CLDC at 1 day after infection, suggesting additional HSV antigens may contribute to protection.
Example 4
[00102] To further evaluate the mechanism of adjuvant activity, mice were vaccinated with gD2 (full-length ectodomain) alone or with gD2 (full-length ectodomain) + CLDC.
Splenocytes from mice vaccinated with gD2 alone, gD2 + CLDC, or control splenocytes were re-stimulated with gD2 antigen and supernatants were collected after 96 hours and assayed by ELISA for murine IFN-γ. Higher levels of total anti-gD2 IgG antibody was detected in the group immunized with gD2 + CLDC compared to gD2 alone (PO.01) (data not shown). In addition, the ratio of IgG2a to IgGl was increased in the gD2 + CLDC group
(data not shown). As shown in Figure 6, splenocytes from mice vaccinated with gD2 +
CLDC showed a significant production of IFN-γ when re-stimulated with gD2 antigen
(P<0.05). In agreement with previously published studies, IFN-γ production in response to gD2 was primarily produced by CD4+ T cells, since depletion of the CD4+ T cell population resulted in a significantly lower level of IFN-γ (BenMohamed L, Bertrand G, McNamara CD,
Gras-Masse H, Hammer J, Wechsler SL, and AB Nesburn. 2003. Identification of novel immunodominant CD4+ Thl-type T cell peptide epitopes from herpes simplex glycoprotein
D that confer protective immunity. J. Virol. 77: 9463, Bettahi I, Zhang X, Afifi RE, and L
BenMohamed. 2006. Protective immunity to genital herpes simplex virus type 1 and type 2 provided by self-adjuvanting lipopeptides that drive dendritic cell maturation and elicit a polarized Thl immune response. Viral Immunol. 19(2):220.). In contrast, CD8+ depleted splenocytes produced similar levels of IFN-γ as shown in Figure 6 (data not shown).
[00103] Thus, gD2 + CLDC vaccine promoted a superior Thl antibody and T cell response in mice and that it was at least equivalent to gD2 + MPL in protecting mice from HSV-2 infection.
Example 5
[00104] Because the guinea pig model also allows evaluation of the severity of acute disease, clinical recurrences, recurrent vaginal shedding and latent virus levels in the dorsal root ganglia, it is considered by many the small animal of choice for evaluation of vaccines. Therefore, the effects of vaccination with gD2 (full-length ectodomain) + CLDC were compared to gD2 (full-length ectodomain) + MPL/alum in the guinea pig model of genital herpes. The gD2 + MPL/alum mimics the vaccine that was recently evaluated in two large clinical trials and is currently being further evaluated in a third clinical trial.
[00105] To begin the studies, sixty guinea pigs were randomized into five groups (N=12/group): Group 1 : no vaccine; Group 2: CLDC alone; Group 3: gD2 alone: Group 4: gD2 + CLDC; and Group 5: gD2 + MPL/alum. Animals were immunized on days 49 and 21 days prior to viral inoculation. Each dose was administered by subcutaneous injection at five separate sites on the dorsum. One day before viral challenge, animals were bled by toenail clip and the serum stored at -20°C. The studies found, specific antibody to gD2 was increased by the combination of gD2 with both the CLDC and MPL/alum adjuvants (data not shown).
[00106] Animals were inoculated intravaginally with 5x105 pfu HSV-2, strain MS similar to previous studies (Bernstein DI, Harrison CJ, Jenski LJ, Myers MG, and LR Stanberry. 1991. Cell-mediated immunologic responses and recurrent genital herpes in the guinea pig: effects of glycoprotein immunotherapy. J. Immunol. 146:3571.). Swab samples of cervicovaginal secretions were collected on 1, 2, 4, 6, 8 and 10 days post infection (dpi) and stored frozen (-70°C) until assayed for virus titers by plaque assay. Guinea pigs were evaluated daily and primary genital skin disease was quantified using a lesion score-scale ranging from 0, representing no disease, to 4, representing severe vesiculoulcerative skin disease of the perineum. Following recovery from primary infection, animals were examined daily from 21-63 dpi for evidence of spontaneous recurrent herpetic lesions. The number of lesion days (days on which a recurrent lesion was observed on the perineum) was used as a measure of the frequency of recurrent disease. Vaginal swabs also were obtained every three days from days 21-63 to evaluate for recurrent virus shedding. Swabs were stored frozen (-70°C) until they were processed for PCR analysis to determine the frequency and magnitude of viral shedding into the genital tract. At the end of the study, the guinea pigs were sacrificed, and the DRGs were harvested aseptically. DRGs were stored frozen (-80°C) until DNA was extracted by PCR for determination of the magnitude of latent virus infection.
[00107] As shown in Figure 7, following vaccination at 21 and 49 days prior to virus challenge, the gD2 + CLDC vaccine significantly decreased the severity of genital disease compared to vaccine alone (P<0.001) and compared to the gD2 + MPL/alum vaccine
(P=0.060). The only animals that did not develop disease were in the groups receiving gD2 + CLDC (5 animals or 42%) and the group receiving gD2 + MPL/alum (2 animals or 17%).
[00108] As shown in Figure 8, only vaccination with an adjuvant significantly decreased the amount of virus shed during the acute disease. On 4 and 6 dpi, the amount of virus shed in the group receiving gD2 + CLDC was significantly less than the group receiving gD2 alone and likewise, the gD2 + CLDC shed significantly less virus on 2 and 4 dpi than the group receiving gD2 + MPL/alum (P<0.05).
[00109] The effects of vaccination on subsequent recurrent disease are shown in
Figure 9. The data are depicted as cumulative mean lesion days from 3 to 9 weeks after challenge. Most importantly, the number of days with recurrences was significantly less in the gD2 + CLDC group (0.4 ± 1.4) compared to gD2 alone (2.8 ± 3.6) or gD2 + MPL/alum group (2.0 ± 2.4). PO.0001 for gD2+CLDC versus no vaccine; PO.008 for gD2 +
MPL/alum versus no vaccine.
[00110] As shown in Figure 10, vaccination with gD2 + CLDC, or gD2 + MPL/alum significantly decreased the number of animals developing recurrences compared to the gD2 alone group (PO.001 and PO.07, respectively). The gD2 + CLDC also had significantly fewer animals with subsequent recurrent disease compared to the gD2 alone group (P<0.001), whereas the gD2 + MPL/alum group was not significantly different. Notably, only one out of 12 animals (8%) in the gD2 + CLDC group developed subsequent recurrent lesions compared to six out of 12 (50%) for the gD2 + MPL/alum group (p=0.07). Similarly, the mean number of days with recurrent lesions was decreased by immunization, as shown in Figure 11. As shown in Figure 12, the number of animals that shed virus after the acute disease had resolved was not significantly different between groups. However, only vaccination with gD2 + CLDC reduced the number of days with recurrent virus shedding to 2.2 ± 1.3 days in the gD2 + CLDC compared to 3.5 ± 1.4 days in the gD2 alone group and 4.7 ±1.6 days in gD2 + MPL/alum (PO.05 for each comparison to CLDC). Example 6
[00111] Various HSV-2 subunit vaccines were introduced into guinea pigs (12 guinea pigs/group) with the following groups: 1) no vaccine (10% sucrose only); 2) gD-FL (full length ectodomain of gD2, amino acids 1-306) + CLDC; 3) gD-T (truncated gD2 amino acids 1-285) + CLDC; 4) gD-T + gB2t (amino acids 31-726) + gH2t/gL2 (amino acids 21-802) + CLDC; and 5) gB2 + gH2/gL2) +CLDC. The guinea pigs were immunized subcutaneously twice at three-week intervals, and challenged intravaginally three weeks later with lxl 06 pfu (total) HSV-2 (MS strain). All of the vaccinated groups had significantly reduced acute lesions and virus titers at 2 and 5 dpi compared to the no vaccine group (p<0.002); whereas no significant differences were observed between the vaccinated groups.
[00112] Vaccination with any of the vaccines significantly reduced recurrent HSV-2 disease (p< 0.002) compared to the no vaccine group. For all of the vaccination groups, recurrent mean lesion scores (~1.0 versus 7.5) and recurrent mean lesion days (~1 day versus 9 days) were significantly reduced compared to the no vaccine group (Figures 14-15). Swab samples were collected on 1, 2, 5 and 8 days(dpi) and assayed for virus titers. The animals were scored for primary disease for days 3-14 and for recurrent disease for days 15-60.
[00113] PCR analysis of latent viral DNA in the DRG after 63 dpi demonstrated a significant difference (p <0.01) following first round PCR Groups 3, 4, and 5 compared to Group 1 , while Group 1 was not significantly different. Following nested PCR, all groups had 80% of the DRGs positive for HSV-2 DNA, suggesting subtle differences between latent viral load among the vaccination groups. Data shown below and in Figure 13.
Groups # Pos 1st 100ng # Pos 2nd 100ng # Pos 250ng
No Vaccine 9 75.00% 11 91.67% 12 100.00% gD-FL +CLDC 7 58.33% 12 100.00% 8 66.67% gD-T + CLDC 2 16.67% 11 91.67% 6 50.00% gD-T + (gB+gH/gL) + CLDC 1 8.33% 10 83.33% 9 75.00%
(gB+gH/gL) + CLDC 2 16.67% 11 91.67% 12 100.00% Example 7
[00114] Various preparations of cationic liposome delivery vehicles, such as JVRS- 100, are described in U.S. Patent 6,693,086 and as follows. For example, a cationic liposome consisting of DOTAP (1,2 dioleoyl-3-trimethylammonium-propane) and cholesterol was mixed in a 1 :1 molar ratio, dried down in round bottom tubes, and then rehydrated in 5% dextrose solution (D5W) by heating at 50° C for 6 hours, as described previously (Solodin et al., 1995, Biochemistry, 34: 13537-13544). This procedure results in the formation of liposomes that consists of multilamellar vesicles (MLV), which provide optimal transfection efficiency when compared to small unilamellar vesicles (SUV). The production of MLVs and related "extruded lipids" is also described in Liu et al., 1997, Nature Biotech., 15: 167-173; and Templeton et al., 1997, Nature Biotech.. 15:647-652.
Example 8
[00115] Sf9 cells are grown to a density of 4xl06 cells per mL in a Celligen bioreactor or Spinner vessel and infected with a recombinant virus at an MOI of 4. The cell supernatant is separated from cells when the cell viability drops to 70 percent (typically, 72 hours postinfection). Proteins are purified by one or two different methods:
[00116] 1. Immunoaffinity chromatography. As examples, gD2(306t) and gD2(285t) are purified from the culture supernatant by immunoaffinity chromatography using the gD type-common monoclonal antibody, DL6, a non-neutralizing antibody that recognizes a continuous epitope at 272-279. The procedure is identical to that used for gDl(306t) (Sisk, W. P., J. D. Bradley, et al. (1994). "High-level expression and purification of secreted forms of herpes simplex virus type 1 glycoprotein gD synthesized by baculovirus-infected insect cells." J. Virol. 68: 766-775.). After washing with a buffer consisting of 10 mM Tris HCl, pH 7.2, and 500 mM NaCl, bound gD is eluted from the column with 3M KSCN. The recovered protein is dialyzed against PBS and concentrated prior to storage at -80°C. A typical recovery of purified gD2 (306t) is 5-10 mg/L of supernatant fluid. DL16, a trimer specific antibody, was used to purify gBl but it binds gB2 poorly.
[00117] 2. Nickel affinity chelate chromato raphy. Some gB and gD constructs were prepared using nickel affinity chelate chromatography. To utilize this procedure, the protein must have an accessible 6-His tag (SEQ ID NO: 17). Briefly, the supernatant fluid from infected Sf9 cells was concentrated, dialyzed and the his-tagged protein was adsorbed onto nickel beads, and washed with a buffer to get rid of contaminating proteins that adhere to the beads. For some proteins, the wash step might just require elevated salt and in other cases, such as for gH/gL, a low concentration of imidazole is needed. This step gets rid of the contaminants, but somewhat lowers the yield. The protein was eluted with imidazole, dialyzed, concentrated and stored at about -80°C. About 1-2 mg of gH/gL2 was obtained per liter of media.
Example 9
[00118] DNA encoding amino acid residues 1-306 (not including the signal sequence) of gD2 from the HSV-2 strain 333 was modified with sequences for a 6-His tag (SEQ ID NO: 17) (after residue 306) was cloned into the baculo virus transfer vector pVT-Bac (Tessier, D. et al., Gene 98: 177-183, 1991). It was cloned in frame with the DNA sequence of the honey bee melittin signal peptide. The resulting plasmid is pAN243.
[00119] Using the same pVT-Bac vector, a further gD2 truncated form (gD2t (285)) was cloned. The resulting plasmid is pYZ343. This construct also has a His tag after residue 285. Also cloned was a "His-less" version of gD2t (285), for which the plasmid is pYZ342. [00120] DNA encoding amino acid residues 31-726 of the glycoprotein gB2 from HSV-2 strain 333 was inserted into the pVT-Bac vector in frame with the melittin signal sequence (pCW289).
[00121] DNA encoding amino acid residues (21-802) of glycoprotein gH2 with a 6-His tag (SEQ ID NO: 17) and full length glycoprotein gL2 (which lacks a transmembrane domain or cytoplasmic tail) from HSV-2 strain 333 were cloned into the vector pFastBacDual (pTC605). This vector allows one to clone two genes in frame with two different promoters. The proteins were cloned to include their native signal sequences.
[00122] A mammalian cell line was constructed, which secretes a soluble gH/gL complex, consisting of gH truncated at amino acid residue 792 (gH-lt792) and full length gL, both from HSV-1. Purified gH-lt792/gLl reacted with gH/gL specific monoclonal antibodies, including LP1 1, indicating that the complex retained its proper antigenic structure. Polyclonal antibodies to the complex did block entry and exhibited high titers of complement-independent neutralizing activity against HSV-1, and showed limited cross- neutralization of HSV-2.
[00123] Mice were immunized with the complex. Using a zosteriform model of infection, mice were challenged with HSV-1. Mice immunized with either gD or gH- lt792/gLl had reduced primary lesions and exhibited no secondary zosteriform lesions and survived virus challenge.
[00124] The gH-lt792/gLl and gH-2t792/gL2 forms were prepared in the baculovirus expressions and then mice were immunized with the two sets of proteins. Both sets of sera neutralized their forms of the virus, but there was no cross-reactivity of gHl/gLl to HSV-2 or gH2/gL2 to HSV-1. [00125] Also prepared were complexes of gD2 with gH2/gL2 wherein the gD2 full length ectodomain or the truncated form, gD-2t (285), were appended onto the N-terminus of gH2. The tripartite complex functioned in infection and in inducing cell-cell fusion. The gD- 2t(285) has a much higher affinity for the receptor than the full-length ectodomain form.
Example 10
[00126] These studies tested the immunogenicity of single and multi-antigen HSV2 subunit vaccines in both mice and non-human primates (cynomolgus monkey). The addition of adjuvant, either MPLalum or JVRS-100 (a cationic lipid-DNA complex) was beneficial for protection from disease progression and death in a mouse HSV2 challenge model regardless of whether it was composed of subunit glycoprotein gD (e.g., gD2t) and/or cellular entry antigens gB, gH and gL. In both the mouse and non-human primate studies, adjuvant increased the antigen specific and neutralizing antibody titers as well as the frequencies of HSV2 specific CD4+ and CD8+ T cell responses as measured by Thl cytokine production. Interestingly in the mouse study, the data showed an inverse relationship between
immunogenicity and increased antigen dose. An advantageous effect of a combinatorial antigen vaccine was shown in the non-human primate study and in the mouse study, but the responses differed most notably in increased T cell responses in the monkey model.
[00127] Animals. Female Balb/c mice approximately 6-8 weeks were obtained from Harlan Laboratories (Indianapolis IN) and group housed meeting the requirements of the United States Department of Agriculture (USDA) and the Guide for the Care and Use of Laboratory Animals. Chinese origin cynomolgus monkeys (Macaca fasicularis) were supplied by Three Springs Scientific (Perkasie PA) and housed in accordance with the regulations of the American Association for Accreditation of Animal Care Standards (AAALAC). At the time of study entry all non-human primates had tested negative for Herpes B antibodies.
[00128] Vaccine preparation/administration. Vaccines were prepared within four hours of vaccination by mixing the relevant antigen and adjuvant components (Tables 1 and 3). The HSV2 antigens were prepared from insect cells infected with a recombinant baculovirus expressing either gD2, gD2t, gB or gH/gL as previously described (Willis SH et al, J. Virol. 72:5937-47, 1998). Monophosphoryl-lipid A (MPL) was purchased from Sigma- Aldrich Corp, St. Louis, MO and resuspended in 0.5% (v/v) triethanolamine in water. The MPL/ Alum combination contained 2.5 μg MPL and l(^g aluminum potassium sulfate (Sigma) for mouse studies or 5(^g MPL and 20C^g aluminum potassium sulfate for non- human primate studies. JVRS-100 (Juvaris BioTherapeutics, Inc., Burlingame, CA) was provided as a white, lyophilized powder manufactured from plasmid DNA complexed with liposomes. The lyophilized CLDC drug product was reconstituted in sterile water for injection. After reconstitution, the final drug product was a colloidal dispersion of 0.3mg/mL DNA, 1.88mg/mL DOTIM and 1.05mg/mL cholesterol at pH 7 containing 1.4mM Tris-HCl and 10% w/v lactose (Fairman J et al, Hum. Vaccin. 5(3): 141-50, 2009). 2^g or 50μg DNA weights of JVRS-100 were used respectively for mouse and non human primate studies. An overview of the mouse and non-human primate vaccine regimens is shown in Tables 1 and 3. All vaccinations were intramuscular with a 3 week period between primary inoculation and booster inoculation.
[00129] HSV2 Challenge. Five out of ten mice per group were designated for HSV2 viral challenge. The designated animals were administered 2mg DEPO-PROVERA depot medroxyprogesterone acetate at a dose volume of O.lmL/animal via subcutaneous injection in the upper dorsal region on study day 31. On study day 36 the same animals received a challenge dose of 5x LD50 HSV2 strain MS intravaginally at a dose volume of lC^L/animal. Vaginal HSV2 infection induced clinical scores were recorded at least once daily for 24 days following viral challenge. Clinical scores were based on a grading scale of 0-5. A score of zero indicated no clinical signs of disease; one, vaginal erythema; two, vaginal erythema and edema; three, vaginal herpetic lesions; four, severe vaginal ulceration and/or hind limb paralysis; and five, death.
[00130] Vaginal Viral Titers. Surviving animals from viral challenge had vaginal swabs collected on days 38, 40 and 42. Swabs were placed in lmL DMEM containing antibiotics, vortexed for 30 seconds in triplicate and divided into aliquots for storage at -70°C until viral titer analysis. On the day prior to performing the assay, 6-well tissue culture plate were seeded with 3-3.5 x 105 Vero cells/2mL DMEM low glucose media fortified with 10% fetal bovine serum (FBS) and lx PenStrep. Plates were used when they reached 80-100% confluence at approximately 16-20 hours post seeding. Ten fold serial dilutions were prepared for each swab sample. Samples were tested at dilutions from zero to 10-5. Dilution volumes of 200 ΐ were applied to cells after media removal and allowed to infect for 90 to 150 minutes in a 37°C incubator. Plates were overlaid with 4mL of warmed media and returned to the 37°C incubator for two days. The cells were stained with 30μΙ 1% Neutral Red and incubated an additional 12 hours before counting plaque numbers for each dilution.
[00131] Serum HSV2 Neutralization Assay. Two-fold serial dilutions of heat- inactivated serum or plasma from 1 :25 to 1 :800 were mixed with an equal volume of virus containing approximately 100 PFU's and incubated at 37°C for one hour. After removal of growth media, each serum-virus mixture was added to 80-90% confluent Vero cells. After a 90 minute 37°C incubation plates were overlaid with EMEM containing 0.5% agarose, 5% FBS and lx PenStrep. Plates were incubated for two days at 37°C and then either stained with 1.5% Neutral Red for an additional 12 hours (mouse study) or fixed with ice-cold acetone (80% v/v in PBS) and stained with crystal violet (non human primate study). Plaques were counted and the end point serum neutralization titers were defined as the final serum dilution that produced a >50% reduction in the number of viral plaques as compared to wells on the same plate containing only virus.
[00132] HSV2 Antigen Specific ELISA. Antibody was measured by ELISA using HSV-2 glycoprotein as the solid phase. High binding plates were coated with 200μί of 0.5mg/ml HSV2 antigen diluted in PBS and after washing, blocking with 1%BSA in PBS and washing again, 5-fold serial dilutions of serum or plasma were applied. Plates were incubated for a minimum of 2 hours before washing and adding HRP-conjugated anti-mouse IgG or anti-monkey IgG (Southern Biotechnology, Birmingham, AL) for detection. The plates were then developed by addition of TMB substrate (ThermoFisher Scientific,
Rockford, IL). Stop solution (1M H2S04) was then added and the absorbance measured at 450nm and 570nm. The reading at 570nm was subtracted from the reading at 450nm to correct for plate abnormalities and bubbles in the analyte solution. The resulting data for each sample was plotted to obtain a curve of the inverse of the dilution versus the A45o-As7o measurement. The antibody titer was calculated as the midpoint of the curve or EC50 calculation using Prism statistical software.
[00133] Flow cytometric measurement of HSV2-specific T-cell responses. Freshly isolated splenocytes or peripheral blood mononuclear cells (PBMC) were assayed for their ability to secrete IFNy, TNFa or IL-2 during in vitro restimulation with HSV2 (strain G) virus. Briefly, cells were stimulated for six hours with virus (1 MOI) or medium alone. Brefeldin-A at l(^g/mL was added to the culture for the final 4 hours of stimulation. After stimulation the cells were surface stained with fluorescently conjugated antibodies to CD3, CD4 and CD8. Subsequently the cells were washed and treated with
Fixation/Permeabilization Solution(s) (eBioscience, SanDiego, CA). Permeabilized cells were stained with fluorescently conjugated antibodies to IFNy, TNFa, and IL-2 washed and then fixed with 2% paraformaldehyde. The antibodies were used in the following panels: Mouse; CD3-FITC, CD4-Alexa 700, CD8-PerCP Cy5.5, IFNy-PE Cy7, TNFa-APC, IL2-PE and Non Human Primate; CD3-Alexa 700, CD4-PerCP Cy5.5, CD8-PE Cy7, IFNy-FITC, TNFa-PE, IL2-APC (BD Biosciences, San Jose, CA). One hundred thousand events were collected on the FACs Canto and the data analyzed with FACs Diva software.
[00134] Mouse HSV2 viral challenge after vaccination. The effect of the various HSV-2 antigens alone and in combination with and without the addition of adjuvants MPLalum or JVRS-100 (cationic lipid DNA complex) was evaluated. Included were the cellular entry subunit glycoproteins gD in a truncated format (i.e., residues 1-285; termed "gD2t"), gB and gH/gL. Animals were vaccinated according to a two dose regimen (Table 1) prior to being subject to intravaginal challenge with HSV-2. Survival and disease score data (Figure 16) underscore the requirement for inclusion of an adjuvant in vaccine formulations. None of the unadjuvanted groups had survival above forty percent whereas of the adjuvanted groups only gH/gL alone had survival below eighty percent. The disease scores patterns show more prolonged and severe symptoms in the unadjuvanted groups. Three groups that were one hundred percent protected from developing herpetic lesions were high dose gD2t plus JVRS-100, gD2t plus MPL/alum and triple combination gD2t, gB and gH/gL plus JVRS-100. Viral titers of vaginal swabs (Figure 17) indicate only the triple combination plus JVRS-100 prevented viral replication after challenge. The challenge data show both the gD2t adjuvanted and combination adjuvanted groups had superior survival, viral titers and disease scores compared to the unadjuvanted gD2t and all gB, gH/gL groups. [00135] Mouse humoral and cellular responses to vaccination. Adujvants enhanced the production of antigen specific antibodies by ten-fold, an average of one log (Figure 18). The ELISA assay shows at least in vitro cross reactivity of anti-gD antibodies with gB but not vice versa. The antigen gB induced the highest antibody titers. Increasing the antigen dose four-fold from ^g to 4μg overall resulted in a modest half-log antibody boost. The addition of multiple antigens did not appear to reduce the individual antibody titers to each specific antigen. Neutralizing antibody evaluation (Table 2) also showed gB to be the most potent antigen with 5 of 5 animals in the gB plus JVRS-100 and 4 of 5 animals in the combination plus JVRS-100 group containing gB, having detectable viral neutralizing titers. According to the cellular studies, adjuvant increased virus-specific CD4 and CD8 response measured by intracellular cytokine staining and flow cytometry (Figure 19). It was determined that ^g gD2t plus adjuvant was better than 4μg with adjuvant at eliciting effective T-cell responses.
[00136] Cynomolgus monkey humoral and cellular responses to vaccination.
Animals were vaccinated in a prime boost regimen (Table 3) and their blood evaluated for immune response. Antigen specific antibody titers were overall lower to those seen in the mouse (Figure 20). JVRS-100 was a more potent adjuvant than MPLalum with titers averaging twice as high in the ELISA assay. The form of gD truncated at amino acid residue 285 (gD2t) elicited higher titers than the gD form truncated at residue 306. Also, despite the apparent lesser production of antigen specific antibodies, a higher number of non-human primates raised detectable viral neutralizing antibody titers most notably in the JVRS-100 adjuvanted groups (Table 4). Interestingly, T-cell responses differed from the mouse as well in that the addition of multiple antigens enhanced Thl cytokines production after in vitro stimulation (Figure 21). [00137] The present study addressed the hypothesis that the addition of JVRS-100, a cationic lipid DNA complex, and the combination of multiple cellular entry antigens would result in a more efficacious vaccine than antigens alone or the current human clinical candidate, gD2 plus MPLalum. The study showed that the triple combination of antigens and gD alone when adjuvanted with JVRS-100 were equivalent in the mouse challenge model except triplecombo+JVRS was the only vaccine to prevent viral replication in first 6 days of disease. JVRS-100 and MPLalum adjuvanted vaccines were equivalent in mouse challenge and antibody studies but JVRS-100 was superior to MPLalum adjuvanted vaccines in monkey antibody studies. Detectable neutralizing antibody was generated in the mouse model in response to vaccination with gB or the triple combination of antigen. The assays examining murine T-cell responses suggested that a larger dose was able to improve the T- cell response when administered alone but mice demonstrated an inverse response when increased antigen was combined with adjuvant. Larger amounts dampened the T-cell response in mice as compared to a lower dose. While not wishing to be bound to any particular theory, this effect may have contributed to the lower T-cell responses seen in the combination vaccine as the total dosage was three quarters of the larger gD2 dose 3μg and 4μg respectively.
Table 1 Mouse Vaccination Regimen
Blood/Spleen HSV-2
Group Route and Vaccination Vaginal Swabs
Vaccine/adj uvant Antigen Dose collection challenge
(N=10) volume s (N=5)
(N=5) (N=5)
I gD2t J μ IM, 0. 1 mL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
II gD2t /JVRS-100 1 g IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
III gD2t 4 μg IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
IV gD2t /JVRS-100 4Mg IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
V gD2t / MPLalum ^g IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
VI gB 1 g IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
VII gB/JVRS-100 i μg IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
VIII gHgL i μg IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
IX gHgL/JVRS-100 i μg IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
X gD2t+gB+gHgL 1 μg of ea. IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
XI gD2t+gB+gHgL / JVRS-100 1 g of ea. IM, O. lmL Days 0, 21 Day 36 Day 36 Days 38, 40, 42
XII No treatment N/A N/A Days 0, 21 Day 36 Day 36 Days 38, 40, 42
Table 2 Mouse HSV2 Neutralizing Antibodies
Animals with detectable Average Positive
Group Treatment Neutralizing Antibody Titer
1 gD2t 0/5 N/A
II gD2t /JVRS-100 0/5 N/A
III gD2t 0/5 N/A
IV gD2t /JVRS-100
1/5 50
V gD2t / PLalum 1/5 25
VI gB 0/5 N/A
VII gB/JVRS-100 5/5 60
VIII gH/gL 0/5 N/A
IX gH/gL/JVRS-100 0/5 N/A
X gD2t+gB+gH/gL
1/5 25
XI gD2t+gB+gH/gL / JVRS-100 4/5 25
XII none 0/5 N/A Table 3 Non Human Primate Vaccination Regimen
Antigen Route and Plasma/PBMC
Vaccine/adjuvant Vaccinations
Dose volume collection gD2t / MPLalum 20 μβ IM, 0.5mL Days 0, 21 Day 0, 21, 42 gD2t /JVRS-100 20 μg IM, 0.5mL Days 0, 21 Day O, 21, 42 gD2t+gB+gHgL / 20 μg of ea. IM, 0.5mL Days 0, 21 Day 0, 21, 42 MPLalum
gD2t+gB+gHgL / JVRS 20 μg of ea. IM, 0.5mL Days 0, 21 Day 0, 21, 42
100
gD2t+gB+gHgL 20 μg of ea. IM, 0.5mL Days 0, 21 Day 0, 21, 42 gD2 /JVRS- 100 20 μg IM, 0.5mL Days 0, 21 Day O, 21, 42
Table 4 Non-human Primate HSV2 Neutralizing Antibodies
Animals with detectable Average Positive
Group Treatment Neutralizing Antibody Titer
1 gD2t/ MPLalum 1/4 100
II gD2t/JVRS-100 4/4 68.75
III gD2t+gB+gH/gL / MPLalum 0/4 N/A
IV gD2t+gB+gH/gL /JVRS-100 4/4 56.25
V gD2t+gB+gH/gL 1/4 25
VI gD2 /JVRS-100 4/4 100
[00138] The term "comprising", which is used interchangeably with "including," "containing," or "characterized by," is inclusive or open-ended language and does not exclude additional, unrecited elements or method steps. The phrase "consisting of excludes any element, step, or ingredient not specified in the claim. The phrase "consisting essentially of limits the scope of a claim to the specified materials or steps and those that do not materially affect the basic and novel characteristics of the claimed invention. The present disclosure contemplates embodiments of the invention compositions and methods corresponding to the scope of each of these phrases. Thus, a composition or method comprising recited elements or steps contemplates particular embodiments in which the composition or method consists essentially of or consists of those elements or steps.
[00139] All references cited herein, including patents, patent applications, and publications, are hereby incorporated by reference in their entireties, whether previously specifically incorporated or not.
[00140] Having now fully described this invention, it will be appreciated by those skilled in the art that the same can be performed within a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention and without undue experimentation.
[00141] While this invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications. This application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth.
[00142] Although the invention has been described with reference to the above examples, it will be understood that modifications and variations are encompassed within the spirit and scope of the invention. Accordingly, the invention is limited only by the following claims.

Claims

WHAT IS CLAIMED IS:
1. A composition of isolated Herpes Simplex Virus (HSV) antigens comprising an isolated glycoprotein B (gB) or a fragment thereof, and an isolated glycoprotein H (gH) or a fragment thereof in a complex with glycoprotein L (gL) of a fragment thereof
2. The composition of Claim 1, further comprising an isolated glycoprotein D (gD) or fragment thereof.
3. The composition of Claim 1 or Claim 2, wherein the glycoprotein B is glycoprotein B2 (gB2), the glycoprotein H is glycoprotein H2 (gH2), the glycoprotein L is glycoprotein L2 (gL2), and the glycoprotein D is glycoprotein D2 (gD2).
4. The composition of Claim 1 or Claim 2 further comprising an adjuvant.
5. The composition of Claim 4 wherein the adjuvant is a cationic liposome DNA complex (CLDC).
6. The composition of Claim 5 wherein the CLDC comprises a cationic liposome delivery vehicle and an isolated nucleic acid molecule.
7. The composition of Claim 6, wherein the isolated nucleic acid molecule is selected from the group consisting of i) an isolated nucleic acid molecule that is not operatively linked to a transcription control sequence, ii) an isolated bacterially-derived nucleic acid vector without a gene insert, iii) an isolated nucleic acid molecule comprising a non-coding nucleic acid sequence; iv) an isolated recombinant nucleic acid molecule encoding an immunogen operatively linked to a transcription control sequence, and iv) an oligonucleotide comprising a CpG motif.
8. The composition of Claim 6 wherein the cationic liposome delivery vehicle comprises DOTAP and cholesterol; DOTMA and cholesterol; DDAB and cholesterol; or DOTIM and cholesterol.
9. The composition of Claim 1 or Claim 2, wherein the fragment of gB comprises the ectodomain of the full length protein.
10. The composition of Claim 1 or Claim 2, wherein the fragment of gH comprises the ectodomain of the full length protein.
1 1. The composition of Claim 3, wherein the fragment of gB comprises amino acid residues 31-726 or amino acid residues 31-727 of gB2, and the fragment of gH comprises amino acid residues 21-802 of gH2.
12. The composition of Claim 3, wherein the fragment of gD comprises amino acid residues 1-285 of gD2.
13. The composition of Claim 11, wherein the fragment of gB consists of amino acid residues 31-726 of gB2 or amino acid residues 31-727 of gB2.
14. The composition of Claim 11 , wherein the fragment of gH consists of amino acid residues 21-802 of gH2.
15. The composition of Claim 12, wherein the fragment of gD consists of amino acid residues 1-285 of gD2.
16. A composition according to any of the preceding claims for inducing an immune response against HSV in a mammalian subject.
17. A composition according to any of the preceding claims for vaccinating a mammalian subject against HSV-2.
18. The composition for use according to claim 17, wherein the subject has been diagnosed with an HSV-2 infection.
19. The composition for use according to claim 18, wherein the composition is effective to treat the HSV-2 infection.
20. A method for vaccinating a mammalian subject against HSV comprising administering to the subject an effective amount of the composition of any of claims 1-15.
21. A method for vaccinating a mammalian subject against HSV comprising concurrently or sequentially administering to the subject an effective amount of an isolated glycoprotein B (gB) or a fragment thereof in an adjuvant, and an isolated glycoprotein H (gH) or a fragment thereof in a complex with glycoprotein L (gL) of a fragment thereof, wherein the complex is in an adjuvant.
22. The method of Claim 21 further comprising concurrently or sequentially administering an isolated glycoprotein D (gD) or a fragment thereof in an adjuvant.
23. The method of Claim 21 or Claim 22, wherein the adjuvant is a cationic liposome DNA complex (CLDC).
24. The method of Claim 23, wherein the CLDC comprises a cationic liposome delivery vehicle and an isolated nucleic acid molecule.
25. The method of Claim 24, wherein the isolated nucleic acid molecule is selected from the group consisting of i) an isolated nucleic acid molecule that is not operatively linked to a transcription control sequence, ii) an isolated bacterially-derived nucleic acid vector without a gene insert, iii) an isolated nucleic acid molecule comprising non-coding nucleic acid sequence; iv) an isolated recombinant nucleic acid molecule encoding an immunogen operatively linked to a transcription control sequence, and iv) an oligonucleotide comprising a CpG motif.
26. The method of Claim 24, wherein the cationic liposome delivery vehicle comprises DOTAP and cholesterol; DOTMA and cholesterol; DDAB and cholesterol; or DOTIM and cholesterol.
27. The method of Claim 21 or Claim 22, wherein the glycoprotein B is glycoprotein B2 (gB2), the glycoprotein H is glycoprotein H2 (gH2), the glycoprotein L is glycoprotein L2 (gL2), and the glycoprotein D is glycoprotein D2 (gD2).
28. The method of Claim 21 or Claim 22, wherein the fragment of gB comprises the ectodomain of the full length protein.
29. The method of Claim 21 or Claim 22, wherein the fragment of gH comprises the ectodomain of the full length protein.
30. The method of Claim 27, wherein the fragment of gB comprises amino acid residues 31-726 or amino acid residues 31-727 of gB2, and the fragment of gH comprises amino acid residues 21-802 of gH2.
31. The method of Claim 27, wherein the fragment of gD comprises amino acid residues 1-285 of gD2.
32. The method of Claim 30, wherein the fragment of gB consists of amino acid residues 31-726 of gB2 or amino acid residues 31-727 of gB2.
33. The method of Claim 30, wherein the fragment of gH consists of amino acid residues 21-802 of gH2.
34. The method of Claim 31, wherein the fragment of gD consists of amino acid residues 1-285 of gD2.
35. An isolated nucleic acid molecule consisting essentially of a nucleotide sequence encoding glycoprotein B (gB) or a fragment thereof, in-frame with a nucleotide sequence encoding glycoprotein H (gH) or a fragment thereof, and in-frame with a nucleotide sequence encoding glycoprotein L (gL) or a fragment thereof.
36. An isolated nucleic acid molecule consisting essentially of a nucleotide sequence encoding glycoprotein B (gB) or a fragment thereof, in-frame with a nucleotide sequence encoding glycoprotein H (gH) or a fragment thereof, in-frame with a nucleotide sequence encoding glycoprotein L (gL) or a fragment thereof, and in-frame with a nucleotide sequence encoding glycoprotein D (gD) or a fragment thereof.
37. A composition comprising an isolated nucleic acid molecule encoding a glycoprotein B (gB) or a fragment thereof, and an isolated nucleic acid molecule encoding a glycoprotein H (gH) or a fragment thereof, and an isolated nucleic acid molecule encoding glycoprotein L (gL) or a fragment thereof
38. The composition of claim 37, further comprising an isolated nucleic acid molecule encoding glycoprotein D (gD) or a fragment thereof.
39. The composition of Claim 37 or 38, wherein the isolated nucleic acid molecule encoding a fragment of gB comprises amino acid residues 31-726 of glycoprotein B2 (gB2), and the isolated nucleic acid molecule encoding a fragment of gH comprises amino acid residues 21-802 of glycoprotein H2 (gH2).
40. The composition of claim 38, wherein the isolated nucleic acid molecule encoding a fragment of gD comprises amino acid residues 1-285 of glycoprotein D2 (gD2).
41. The composition of Claim 37 or Claim 38, wherein the nucleic acids are complexed with a cationic liposome delivery vehicle to form a cationic liposome DNA complex (CLDC).
42. The composition of Claim 41, wherein the cationic liposome delivery vehicle comprises DOTAP and cholesterol; DOTMA and cholesterol; DDAB and cholesterol; or DOTIM and cholesterol.
43. A method for vaccinating a mammalian subject against HSV-2 comprising concurrently or sequentially administering to the subject an effective amount of an isolated nucleic acid molecule encoding a glycoprotein B (gB) or fragment thereof complexed with a cationic liposome delivery vehicle, and an isolated nucleic acid molecule encoding a glycoprotein H (gH) or fragment thereof in combination with an isolated nucleic acid molecule encoding a glycoprotein L (gL) or fragment thereof, wherein the combination is complexed with a cationic liposome delivery vehicle.
44. The method of Claim 43 further comprising concurrently or sequentially administering an isolated nucleic acid molecule encoding a glycoprotein D (gD) in an adjuvant.
45. The method of Claim 43 or Claim 44, wherein the cationic liposome delivery vehicle comprises DOTAP and cholesterol; DOTMA and cholesterol; DDAB and cholesterol; or DOTIM and cholesterol.
46. The composition of any one of Claims 1-19 and 35-42, further comprising a pharmaceutically acceptable excipient.
47. The composition of Claim 46, wherein the excipient comprises a non-ionic diluent.
48. The composition of Claim 46, wherein the excipient is 5 percent dextrose in water.
PCT/US2011/026188 2010-02-26 2011-02-25 Subunit vaccines for herpes viruses and methods of use WO2011106607A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/578,976 US9089537B2 (en) 2010-02-26 2011-02-25 Subunit vaccines for herpes viruses and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US30885610P 2010-02-26 2010-02-26
US61/308,856 2010-02-26

Publications (2)

Publication Number Publication Date
WO2011106607A2 true WO2011106607A2 (en) 2011-09-01
WO2011106607A3 WO2011106607A3 (en) 2011-10-27

Family

ID=44168478

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/026188 WO2011106607A2 (en) 2010-02-26 2011-02-25 Subunit vaccines for herpes viruses and methods of use

Country Status (2)

Country Link
US (1) US9089537B2 (en)
WO (1) WO2011106607A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017070623A1 (en) * 2015-10-22 2017-04-27 Modernatx, Inc. Herpes simplex virus vaccine
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
WO2018170256A1 (en) * 2017-03-15 2018-09-20 Modernatx, Inc. Herpes simplex virus vaccine
RU2732120C2 (en) * 2015-02-11 2020-09-11 Альма Матер Студиорум Университа Ди Болонья Herpes virus with a modified spectrum of targets containing hybrid glycoprotein h
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
WO2022074358A1 (en) * 2020-10-08 2022-04-14 Virokine Therapeutics Limited Vaccine compositions

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ597182A (en) 2009-05-22 2014-07-25 Genocea Biosciences Inc Vaccines against herpes simplex virus type 2: compositions and methods for eliciting an immune response
WO2012074881A2 (en) * 2010-11-24 2012-06-07 Genocea Biosciences, Inc. Vaccines against herpes simplex virus type 2: compositions and methods for eliciting an immune response
EP2782597B1 (en) * 2011-11-23 2022-04-13 Genocea Biosciences, Inc. Nucleic acid vaccines against herpes simplex virus type 2: compositions and methods for eliciting an immune response
MX355469B (en) 2012-07-06 2018-04-19 Novartis Ag Complexes of cytomegalovirus proteins.
WO2018064232A1 (en) 2016-09-28 2018-04-05 Genocea Biosciences, Inc. Methods and compositions for treating herpes
EP3573649A4 (en) * 2017-01-27 2020-12-30 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Vaccine compositions of herpesvirus envelope protein combinations to induce immune response
WO2018200613A1 (en) * 2017-04-26 2018-11-01 Merck Sharp & Dohme Corp. Hsv antigenic peptides and hsv protein vaccines
WO2023283601A1 (en) * 2021-07-07 2023-01-12 Rational Vaccines, Inc. Vaccines for intracellular pathogens and methods of production

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4762708A (en) 1982-02-18 1988-08-09 University Patents, Inc. Materials and methods for herpes simplex virus vaccination
US5149660A (en) 1982-02-18 1992-09-22 University Patents, Inc. Diagnostic reagents relating to herpes simplex virus
US5244792A (en) 1984-04-06 1993-09-14 Chiron Corporation Expression of recombinant glyoprotein B from herpes simplex virus
US5654174A (en) 1995-07-07 1997-08-05 Competitive Technologies, Inc. Herpes simplex virus glycoprotein D variants
US5807557A (en) 1994-07-25 1998-09-15 The Trustees Of The University Of Pennsylvania Soluble herpesvirus glycoprotein complex
US6156319A (en) 1994-07-25 2000-12-05 The Trustees Of The University Of Pennsylvania Soluble herpesvirus glycoprotein complex vaccine
US6693086B1 (en) 1998-06-25 2004-02-17 National Jewish Medical And Research Center Systemic immune activation method using nucleic acid-lipid complexes

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2257306B1 (en) * 2008-03-25 2014-05-07 Juvaris Biotherapeutics, Inc. Enhancement of an immune response by administration of a cationic lipid-dna complex (cldc)
EP2456463A4 (en) * 2009-07-24 2013-12-04 Merck Sharp & Dohme Recombinant ectodomain expression of herpes simplex virus glycoproteins in yeast

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4762708A (en) 1982-02-18 1988-08-09 University Patents, Inc. Materials and methods for herpes simplex virus vaccination
US5149660A (en) 1982-02-18 1992-09-22 University Patents, Inc. Diagnostic reagents relating to herpes simplex virus
US5244792A (en) 1984-04-06 1993-09-14 Chiron Corporation Expression of recombinant glyoprotein B from herpes simplex virus
US5648079A (en) 1984-04-06 1997-07-15 Chiron Corporation Herpes simplex virus glycoprotein B vaccine
US5759814A (en) 1984-04-06 1998-06-02 Chiron Corporation Expression of recombinant glycoprotein B from herpes simplex virus
US5807557A (en) 1994-07-25 1998-09-15 The Trustees Of The University Of Pennsylvania Soluble herpesvirus glycoprotein complex
US6156319A (en) 1994-07-25 2000-12-05 The Trustees Of The University Of Pennsylvania Soluble herpesvirus glycoprotein complex vaccine
US6541459B1 (en) 1994-07-25 2003-04-01 The Trustees Of The University Of Pennsylvania Soluble herpesvirus glycoprotein complex vaccine
US5654174A (en) 1995-07-07 1997-08-05 Competitive Technologies, Inc. Herpes simplex virus glycoprotein D variants
US5814486A (en) 1995-07-07 1998-09-29 Competitive Technologies, Inc. Herpes simplex virus glycoprotein D variants
US6693086B1 (en) 1998-06-25 2004-02-17 National Jewish Medical And Research Center Systemic immune activation method using nucleic acid-lipid complexes

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
AHMAD A ET AL., J VIROL., vol. 74, no. 16, 2000, pages 7196 - 7203
AURELIAN L ET AL., J GEN VIROL., vol. 68, 1987, pages 2831
BENMOHAMED L; BERTRAND G; MCNAMARA CD; GRAS-MASSE H; HAMMER J; WECHSLER SL; AB NESBURN: "Identification of novel immunodominant CD4+ Thl-type T cell peptide epitopes from herpes simplex glycoprotein D that confer protective immunity", J. VIROL., vol. 77, 2003, pages 9463
BERNSTEIN DI; HARRISON CJ; JENSKI LJ; MYERS MG; LR STANBERRY: "Cell-mediated immunologic responses and recurrent genital herpes in the guinea pig: effects of glycoprotein immunotherapy", J. IMMUNOL., vol. 146, 1991, pages 3571
BETTAHI I; ZHANG X; AFIFI RE; L BENMOHAMED.: "Protective immunity to genital herpes simplex virus type 1 and type 2 provided by self-adjuvanting lipopeptides that drive dendritic cell maturation and elicit a polarized Thl immune response", VIRAL IMMUNOL., vol. 19, no. 2, 2006, pages 220
CUNNINGHAM AL ET AL., J CLIN INVEST., vol. 75, 1985, pages 226
CUNNINGHAM AL ET AL., J CLIN INVESTIG., vol. 83, 1989, pages 490
FAIRMAN J ET AL., HUM. VACCIN., vol. 5, no. 3, 2009, pages 141 - 50
FRIEDMAN HM ET AL., J IMMUNOL., vol. 165, 2000, pages 4528
JUGOVIC P ET AL., J VIROL., vol. 72, 1998, pages 5076
KOELLE DM ET AL., J IMMUNOL., vol. 166, 2001, pages 4049
KOELLE ET AL., J CLIN INVEST., vol. 101, 1998, pages 1500
KOKUBA H ET AL., J INVESTIG DERMATOL., vol. 113, 1999, pages 808
KRUMMENACHER ET AL., EMBO JOURNAL, vol. 24, 2005, pages 4144 - 4153
LIU ET AL., NATURE BIOTECH., vol. 15, 1997, pages 167 - 173
MIKLOSKA Z ET AL., J GEN VIROL., vol. 79, 1998, pages 353
MILLIGAN GN ET AL., J IMMUNOL., vol. 160, no. 12, 1998, pages 6093
NAGASHUNMUGAM T ET AL., J VIROL., vol. 72, 1998, pages 5351
POLLARA G ET AL., J INFECT DIS., vol. 187, 2003, pages 1513
POSOVAD CM ET AL., PROC NATL ACAD SCI USA., vol. 94, 1997, pages 10289
SACKS SL ET AL., ANTIVIRAL RES., 2004, pages 63S1 - S27
SALIO M ET AL., EUR J IMMUNOL., vol. 29, 1999, pages 3245
SISK, W. P.; J. D. BRADLEY ET AL.: "High-level expression and purification of secreted forms of herpes simplex virus type 1 glycoprotein gD synthesized by baculovirus-infected insect cells", J. VIROL., vol. 68, 1994, pages 766 - 775
SOLODIN ET AL., BIOCHEMISTRY, vol. 34, 1995, pages 13537 - 13544
STRIBLING ET AL., PROC. NATL. ACAD. SCI. USA, vol. 189, 1992, pages 11277 - 11281
TEMPLETON ET AL., NATURE BIOTECH., vol. 15, 1997, pages 647 - 652
TESSIER, D. ET AL., GENE, vol. 98, 1991, pages 177 - 183
TOMAZIN R ET AL., J VIROL., vol. 72, no. 3, 1998, pages 2560
WILLIS SH ET AL., J. VIROL., vol. 72, 1998, pages 5937 - 47

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US10022435B2 (en) 2014-04-23 2018-07-17 Modernatx, Inc. Nucleic acid vaccines
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
RU2732120C2 (en) * 2015-02-11 2020-09-11 Альма Матер Студиорум Университа Ди Болонья Herpes virus with a modified spectrum of targets containing hybrid glycoprotein h
WO2017070623A1 (en) * 2015-10-22 2017-04-27 Modernatx, Inc. Herpes simplex virus vaccine
WO2018170256A1 (en) * 2017-03-15 2018-09-20 Modernatx, Inc. Herpes simplex virus vaccine
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
WO2022074358A1 (en) * 2020-10-08 2022-04-14 Virokine Therapeutics Limited Vaccine compositions

Also Published As

Publication number Publication date
US9089537B2 (en) 2015-07-28
WO2011106607A3 (en) 2011-10-27
US20130171234A1 (en) 2013-07-04

Similar Documents

Publication Publication Date Title
US9089537B2 (en) Subunit vaccines for herpes viruses and methods of use
JP3530526B2 (en) Herpes simplex vaccine comprising HSV glycoprotein gD and 3 deacylated monophosphoryl lipid A
US9161973B2 (en) Compositions and methods for vaccinating against HSV-2
EP2184067B1 (en) Severe acute respiratory syndrome DNA vaccine compositions and methods of use
EP2432504B1 (en) Vaccines against herpes simplex virus type 2: compositions and methods for eliciting an immune response
EA034702B1 (en) Liposomal compositions
WO2011112717A1 (en) A novel mucosal vaccination approach for herpes simplex virus type-2
CA3166256A1 (en) Human papillomavirus (hpv) virus-like particles (vlps) vaccine
Tirabassi et al. A mucosal vaccination approach for herpes simplex virus type 2
Nakao et al. Immunotherapy of acute and recurrent herpes simplex virus type 2 infection with an adjuvant-free form of recombinant glycoprotein D-interleukin-2 fusion protein
Manservigi et al. Immunotherapeutic activity of a recombinant combined gB–gD–gE vaccine against recurrent HSV-2 infections in a guinea pig model
JP6684309B2 (en) Therapeutic vaccine for treating herpes simplex virus type 2 infection
US20090004203A1 (en) Methods of treating measles infectious disease in mammals
RU2775424C2 (en) Immunogenic composition of human cytomegalovirus
Zhao et al. Development of COVID-19 Recombinant Subunit Vaccine and Adjuvant
ES2330182B1 (en) VACCINE AGAINST THE HUMAN PAPILOMA VIRUS.
CN116474086A (en) Vaccine reagent for preventing and/or treating herpes zoster
AU2014200035A1 (en) Compositions and methods for vaccinating against HSV-2

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11707301

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 13578976

Country of ref document: US

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 04/01/2013)

122 Ep: pct application non-entry in european phase

Ref document number: 11707301

Country of ref document: EP

Kind code of ref document: A2